51
|
Mollenhauer B, Bowman FD, Drake D, Duong J, Blennow K, El-Agnaf O, Shaw LM, Masucci J, Taylor P, Umek RM, Dunty JM, Smith CL, Stoops E, Vanderstichele H, Schmid AW, Moniatte M, Zhang J, Kruse N, Lashuel HA, Teunissen C, Schubert T, Dave KD, Hutten SJ, Zetterberg H. Antibody-based methods for the measurement of α-synuclein concentration in human cerebrospinal fluid - method comparison and round robin study. J Neurochem 2018; 149:126-138. [PMID: 30125936 PMCID: PMC6587944 DOI: 10.1111/jnc.14569] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/06/2018] [Accepted: 08/15/2018] [Indexed: 01/12/2023]
Abstract
α‐Synuclein is the major component of Lewy bodies and a candidate biomarker for neurodegenerative diseases in which Lewy bodies are common, including Parkinson's disease and dementia with Lewy bodies. A large body of literature suggests that these disorders are characterized by reduced concentrations of α‐synuclein in cerebrospinal fluid (CSF), with overlapping concentrations compared to healthy controls and variability across studies. Several reasons can account for this variability, including technical ones, such as inter‐assay and inter‐laboratory variation (reproducibility). We compared four immunochemical methods for the quantification of α‐synuclein concentration in 50 unique CSF samples. All methods were designed to capture most of the existing α‐synuclein forms in CSF (‘total’ α‐synuclein). Each of the four methods showed high analytical precision, excellent correlation between laboratories (R2 0.83–0.99), and good correlation with each other (R2 0.64–0.93), although the slopes of the regression lines were different between the four immunoassays. The use of common reference CSF samples decreased the differences in α‐synuclein concentration between detection methods and technologies. Pilot data on an immunoprecipitation mass spectrometry (IP‐MS) method is also presented. Our results suggest that the four immunochemical methods and the IP‐MS method measure similar forms of α‐synuclein and that a common reference material would allow harmonization of results between immunoassays. ![]()
Collapse
Affiliation(s)
- Brit Mollenhauer
- Paracelsus-Elena-Klinik, Kassel, Germany.,Department of Neurology, University Medical Center, Goettingen, Germany
| | - Frederick DuBois Bowman
- Department of Biostatistics, Columbia University, Mailman School of Public Health, New York City, New York, USA
| | - Daniel Drake
- Department of Biostatistics, Columbia University, Mailman School of Public Health, New York City, New York, USA
| | - Jimmy Duong
- Department of Biostatistics, Columbia University, Mailman School of Public Health, New York City, New York, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Omar El-Agnaf
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), and College of Science and Engineering, HBKU, Education City, Qatar Foundation, Doha, Qatar
| | - Leslie M Shaw
- Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, Institute on Aging, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | - Adrian W Schmid
- Proteomics Core Facility, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Marc Moniatte
- Proteomics Core Facility, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Jing Zhang
- University of Washington, Seattle, WA, USA
| | - Niels Kruse
- Institute of Neuropathology, University Medical Center, Goettingen, Germany
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, Institute of Physics of Biological Systems, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | | | | | - Kuldip D Dave
- Michael J. Fox Foundation for Parkinson's Research, New York City, New York, USA
| | - Samantha J Hutten
- Michael J. Fox Foundation for Parkinson's Research, New York City, New York, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Molecular Neuroscience, UCL Institute of Neurology, Queens Square, London, UK.,UK Dementia Research Institute at UCL, London, UK
| |
Collapse
|
52
|
Dhouafli Z, Cuanalo-Contreras K, Hayouni EA, Mays CE, Soto C, Moreno-Gonzalez I. Inhibition of protein misfolding and aggregation by natural phenolic compounds. Cell Mol Life Sci 2018; 75:3521-3538. [PMID: 30030591 PMCID: PMC11105286 DOI: 10.1007/s00018-018-2872-2] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 06/12/2018] [Accepted: 07/09/2018] [Indexed: 12/17/2022]
Abstract
Protein misfolding and aggregation into fibrillar deposits is a common feature of a large group of degenerative diseases affecting the central nervous system or peripheral organs, termed protein misfolding disorders (PMDs). Despite their established toxic nature, clinical trials aiming to reduce misfolded aggregates have been unsuccessful in treating or curing PMDs. An interesting possibility for disease intervention is the regular intake of natural food or herbal extracts, which contain active molecules that inhibit aggregation or induce the disassembly of misfolded aggregates. Among natural compounds, phenolic molecules are of particular interest, since most have dual activity as amyloid aggregation inhibitors and antioxidants. In this article, we review many phenolic natural compounds which have been reported in diverse model systems to have the potential to delay or prevent the development of various PMDs, including Alzheimer's and Parkinson's diseases, prion diseases, amyotrophic lateral sclerosis, systemic amyloidosis, and type 2 diabetes. The lower toxicity of natural compounds compared to synthetic chemical molecules suggest that they could serve as a good starting point to discover protein misfolding inhibitors that might be useful for the treatment of various incurable diseases.
Collapse
Affiliation(s)
- Zohra Dhouafli
- Université de Tunis El Manar, Faculté des Sciences de Tunis, 2092, Tunis, Tunisia
- Laboratory of Aromatic and Medicinal Plants, Center of Biotechnology of Borj-Cédria, BP 901, 2050, Hammam-Lif, Tunisia
| | - Karina Cuanalo-Contreras
- The Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX, 77030, USA
| | - El Akrem Hayouni
- Laboratory of Aromatic and Medicinal Plants, Center of Biotechnology of Borj-Cédria, BP 901, 2050, Hammam-Lif, Tunisia
| | - Charles E Mays
- The Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Claudio Soto
- The Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX, 77030, USA
| | - Ines Moreno-Gonzalez
- The Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX, 77030, USA.
- Department of Cell Biology, Networking Research Center on Neurodegenerative Diseases (CIBERNED), Facultad Ciencias, Universidad de Malaga, Málaga, Spain.
| |
Collapse
|
53
|
Olanow CW, Kordower JH. Targeting α-Synuclein as a therapy for Parkinson's disease: The battle begins. Mov Disord 2018; 32:203-207. [PMID: 28218461 DOI: 10.1002/mds.26935] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 01/04/2017] [Accepted: 01/05/2017] [Indexed: 01/13/2023] Open
Affiliation(s)
- C Warren Olanow
- Departments of Neurology and Neuroscience, Mount Sinai School of Medicine, New York 10029, New York, USA.,Clintrex LLC
| | - Jeffrey H Kordower
- Department of Neurological Sciencs, Rush University Schoolf of Medicine, Chicago, Illinois, USA.,Van Andel Research Institute, Grands Rapids Michigan 60612, USA
| |
Collapse
|
54
|
Manoutcharian K, Perez-Garmendia R, Gevorkian G. Recombinant Antibody Fragments for Neurodegenerative Diseases. Curr Neuropharmacol 2018; 15:779-788. [PMID: 27697033 PMCID: PMC5771054 DOI: 10.2174/1570159x01666160930121647] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 09/04/2016] [Accepted: 09/28/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Recombinant antibody fragments are promising alternatives to full-length immunoglobulins and offer important advantages compared with conventional monoclonal antibodies: extreme specificity, higher affinity, superior stability and solubility, reduced immunogenicity as well as easy and inexpensive large-scale production. OBJECTIVE In this article we will review and discuss recombinant antibodies that are being evaluated for neurodegenerative diseases in pre-clinical models and in clinical studies and will summarize new strategies that are being developed to optimize their stability, specificity and potency for advancing their use. METHODS Articles describing recombinant antibody fragments used for neurological diseases were selected (PubMed) and evaluated for their significance. RESULTS Different antibody formats such as single-chain fragment variable (scFv), single-domain antibody fragments (VHHs or sdAbs), bispecific antibodies (bsAbs), intrabodies and nanobodies, are currently being studied in pre-clinical models of cancer as well as infectious and autoimmune diseases and many of them are being tested as therapeutics in clinical trials. Immunotherapy approaches have shown therapeutic efficacy in several animal models of Alzheimer´s disease (AD), Parkinson disease (PD), dementia with Lewy bodies (DLB), frontotemporal dementia (FTD), Huntington disease (HD), transmissible spongiform encephalopathies (TSEs) and multiple sclerosis (MS). It has been demonstrated that recombinant antibody fragments may neutralize toxic extra- and intracellular misfolded proteins involved in the pathogenesis of AD, PD, DLB, FTD, HD or TSEs and may target toxic immune cells participating in the pathogenesis of MS. CONCLUSION Recombinant antibody fragments represent a promising tool for the development of antibody-based immunotherapeutics for neurodegenerative diseases.
Collapse
Affiliation(s)
- Karen Manoutcharian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), Mexico DF. Mexico
| | - Roxanna Perez-Garmendia
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), Mexico DF. Mexico
| | - Goar Gevorkian
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico (UNAM), Apartado Postal 70228, Cuidad Universitaria, Mexico DF, CP 04510, Mexico. 0
| |
Collapse
|
55
|
|
56
|
Bittar A, Sengupta U, Kayed R. Prospects for strain-specific immunotherapy in Alzheimer's disease and tauopathies. NPJ Vaccines 2018; 3:9. [PMID: 29507776 PMCID: PMC5829136 DOI: 10.1038/s41541-018-0046-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 01/17/2018] [Accepted: 01/19/2018] [Indexed: 12/20/2022] Open
Abstract
With increasing age, as the incidence of Alzheimer's disease is increasing, finding a therapeutic intervention is becoming critically important to either prevent or slow down the progression of the disease. Passive immunotherapy has been demonstrated as a successful way of reducing large aggregates and improving cognition in animal models of both tauopathies and Alzheimer's disease. However, with all the continuous attempts and significant success of immunotherapy in preclinical studies, finding a successful clinical therapy has been a great challenge, possibly indicating a lack of accuracy in targeting the toxic species. Both active and passive immunotherapy approaches in transgenic animals have been demonstrated to have pros and cons. Passive immunotherapy has been favored and many mechanisms have been shown to clear toxic amyloid and tau aggregates and improve memory. These mechanisms may differ depending on the antibodie's' target and administration route. In this regard, deciding on affinity vs. specificity of the antibodies plays a significant role in terms of avoiding the clearance of the physiological forms of the targeted proteins and reducing adverse side effects. In addition, knowing that a single protein can exist in different conformational states, termed as strains, with varying degrees of neurotoxicity and seeding properties, presents an additional level of complexity. Therefore, immunotherapy targeting specifically the toxic strains will aid in developing potential strategies for intervention. Moreover, an approach of combinatorial immunotherapies against different amyloidogenic proteins, at distinct levels of the disease progression, might offer an effective therapy in many neurodegenerative diseases.
Collapse
Affiliation(s)
- Alice Bittar
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555 USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Urmi Sengupta
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555 USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555 USA
- Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555 USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555 USA
- Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, TX 77555 USA
| |
Collapse
|
57
|
Rockenstein E, Ostroff G, Dikengil F, Rus F, Mante M, Florio J, Adame A, Trinh I, Kim C, Overk C, Masliah E, Rissman RA. Combined Active Humoral and Cellular Immunization Approaches for the Treatment of Synucleinopathies. J Neurosci 2018; 38:1000-1014. [PMID: 29246926 PMCID: PMC5783958 DOI: 10.1523/jneurosci.1170-17.2017] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 12/04/2017] [Accepted: 12/06/2017] [Indexed: 12/30/2022] Open
Abstract
Dementia with Lewy bodies, Parkinson's disease, and Multiple System Atrophy are age-related neurodegenerative disorders characterized by progressive accumulation of α-synuclein (α-syn) and jointly termed synucleinopathies. Currently, no disease-modifying treatments are available for these disorders. Previous preclinical studies demonstrate that active and passive immunizations targeting α-syn partially ameliorate behavioral deficits and α-syn accumulation; however, it is unknown whether combining humoral and cellular immunization might act synergistically to reduce inflammation and improve microglial-mediated α-syn clearance. Since combined delivery of antigen plus rapamycin (RAP) in nanoparticles is known to induce antigen-specific regulatory T cells (Tregs), we adapted this approach to α-syn using the antigen-presenting cell-targeting glucan microparticle (GP) vaccine delivery system. PDGF-α-syn transgenic (tg) male and female mice were immunized with GP-alone, GP-α-syn (active humoral immunization), GP+RAP, or GP+RAP/α-syn (combined active humoral and Treg) and analyzed using neuropathological and biochemical markers. Active immunization resulted in higher serological total IgG, IgG1, and IgG2a anti-α-syn levels. Compared with mice immunized with GP-alone or GP-α-syn, mice vaccinated with GP+RAP or GP+RAP/α-syn displayed increased numbers of CD25-, FoxP3-, and CD4-positive cells in the CNS. GP-α-syn or GP+RAP/α-syn immunizations resulted in a 30-45% reduction in α-syn accumulation, neuroinflammation, and neurodegeneration. Mice immunized with GP+RAP/α-syn further rescued neurons and reduced neuroinflammation. Levels of TGF-β1 were increased with GP+RAP/α-syn immunization, while levels of TNF-α and IL-6 were reduced. We conclude that the observed effects of GP+RAP/α-syn immunization support the hypothesis that cellular immunization may enhance the effects of active immunotherapy for the treatment of synucleinopathies.SIGNIFICANCE STATEMENT We show that a novel vaccination modality combining an antigen-presenting cell-targeting glucan particle (GP) vaccine delivery system with encapsulated antigen (α-synuclein) + rapamycin (RAP) induced both strong anti-α-synuclein antibody titers and regulatory T cells (Tregs). This vaccine, collectively termed GP+RAP/α-syn, is capable of triggering neuroprotective Treg responses in synucleinopathy models, and the combined vaccine is more effective than the humoral or cellular immunization alone. Together, these results support the further development of this multifunctional vaccine approach for the treatment of synucleinopathies, such as Parkinson's disease, dementia with Lewy bodies, and multiple systems atrophy.
Collapse
Affiliation(s)
- Edward Rockenstein
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093-0624
| | - Gary Ostroff
- University of Mass Massachusetts Medical School, Program in Molecular Medicine Worcester, Massachusetts 01605
| | - Fusun Dikengil
- University of Mass Massachusetts Medical School, Program in Molecular Medicine Worcester, Massachusetts 01605
| | - Florentina Rus
- University of Mass Massachusetts Medical School, Program in Molecular Medicine Worcester, Massachusetts 01605
| | - Michael Mante
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093-0624
| | - Jazmin Florio
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093-0624
| | - Anthony Adame
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093-0624
| | - Ivy Trinh
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093-0624
| | - Changyoun Kim
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093-0624
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Cassia Overk
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093-0624
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093-0624
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland 20892, and
| | - Robert A Rissman
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093-0624,
- Veterans Affairs San Diego Healthcare System, La Jolla, California 92161
| |
Collapse
|
58
|
More SV, Choi DK. Emerging preclinical pharmacological targets for Parkinson's disease. Oncotarget 2018; 7:29835-63. [PMID: 26988916 PMCID: PMC5045437 DOI: 10.18632/oncotarget.8104] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 02/08/2016] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurological condition caused by the degeneration of dopaminergic neurons in the basal ganglia. It is the most prevalent form of Parkinsonism, categorized by cardinal features such as bradykinesia, rigidity, tremors, and postural instability. Due to the multicentric pathology of PD involving inflammation, oxidative stress, excitotoxicity, apoptosis, and protein aggregation, it has become difficult to pin-point a single therapeutic target and evaluate its potential application. Currently available drugs for treating PD provide only symptomatic relief and do not decrease or avert disease progression resulting in poor patient satisfaction and compliance. Significant amount of understanding concerning the pathophysiology of PD has offered a range of potential targets for PD. Several emerging targets including AAV-hAADC gene therapy, phosphodiesterase-4, potassium channels, myeloperoxidase, acetylcholinesterase, MAO-B, dopamine, A2A, mGlu5, and 5-HT-1A/1B receptors are in different stages of clinical development. Additionally, alternative interventions such as deep brain stimulation, thalamotomy, transcranial magnetic stimulation, and gamma knife surgery, are also being developed for patients with advanced PD. As much as these therapeutic targets hold potential to delay the onset and reverse the disease, more targets and alternative interventions need to be examined in different stages of PD. In this review, we discuss various emerging preclinical pharmacological targets that may serve as a new promising neuroprotective strategy that could actually help alleviate PD and its symptoms.
Collapse
Affiliation(s)
- Sandeep Vasant More
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, South Korea
| | - Dong-Kug Choi
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chungju, South Korea
| |
Collapse
|
59
|
Maurel C, Dangoumau A, Marouillat S, Brulard C, Chami A, Hergesheimer R, Corcia P, Blasco H, Andres CR, Vourc'h P. Causative Genes in Amyotrophic Lateral Sclerosis and Protein Degradation Pathways: a Link to Neurodegeneration. Mol Neurobiol 2018; 55:6480-6499. [PMID: 29322304 DOI: 10.1007/s12035-017-0856-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 12/20/2017] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a disease caused by the degeneration of motor neurons (MNs) leading to progressive muscle weakness and atrophy. Several molecular pathways have been implicated, such as glutamate-mediated excitotoxicity, defects in cytoskeletal dynamics and axonal transport, disruption of RNA metabolism, and impairments in proteostasis. ALS is associated with protein accumulation in the cytoplasm of cells undergoing neurodegeneration, which is a hallmark of the disease. In this review, we focus on mechanisms of proteostasis, particularly protein degradation, and discuss how they are related to the genetics of ALS. Indeed, the genetic bases of the disease with the implication of more than 30 genes associated with familial ALS to date, together with the important increase in understanding of endoplasmic reticulum (ER) stress, proteasomal degradation, and autophagy, allow researchers to better understand the mechanisms underlying the selective death of motor neurons in ALS. It is clear that defects in proteostasis are involved in this type of cellular degeneration, but whether or not these mechanisms are primary causes or merely consequential remains to be clearly demonstrated. Novel cellular and animal models allowing chronic expression of mutant proteins, for example, are required. Further studies linking genetic discoveries in ALS to mechanisms of protein clearance will certainly be crucial in order to accelerate translational and clinical research towards new therapeutic targets and strategies.
Collapse
Affiliation(s)
- C Maurel
- UMR INSERM U1253, Université de Tours, 37032, Tours, France
| | - A Dangoumau
- UMR INSERM U1253, Université de Tours, 37032, Tours, France
| | - S Marouillat
- UMR INSERM U1253, Université de Tours, 37032, Tours, France
| | - C Brulard
- UMR INSERM U1253, Université de Tours, 37032, Tours, France
| | - A Chami
- UMR INSERM U1253, Université de Tours, 37032, Tours, France
| | - R Hergesheimer
- UMR INSERM U1253, Université de Tours, 37032, Tours, France
| | - P Corcia
- UMR INSERM U1253, Université de Tours, 37032, Tours, France
- Service de Neurologie, CHRU de Tours, 37044, Tours, France
| | - H Blasco
- UMR INSERM U1253, Université de Tours, 37032, Tours, France
- Service de Biochimie et de Biologie Moléculaire, CHRU de Tours, 37044, Tours, France
| | - C R Andres
- UMR INSERM U1253, Université de Tours, 37032, Tours, France
- Service de Biochimie et de Biologie Moléculaire, CHRU de Tours, 37044, Tours, France
| | - P Vourc'h
- UMR INSERM U1253, Université de Tours, 37032, Tours, France.
- Service de Biochimie et de Biologie Moléculaire, CHRU de Tours, 37044, Tours, France.
| |
Collapse
|
60
|
Choi YR, Cha SH, Kang SJ, Kim JB, Jou I, Park SM. Prion-like Propagation of α-Synuclein Is Regulated by the FcγRIIB-SHP-1/2 Signaling Pathway in Neurons. Cell Rep 2018; 22:136-148. [DOI: 10.1016/j.celrep.2017.12.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 09/01/2017] [Accepted: 12/04/2017] [Indexed: 11/29/2022] Open
|
61
|
Ke PC, Sani MA, Ding F, Kakinen A, Javed I, Separovic F, Davis TP, Mezzenga R. Implications of peptide assemblies in amyloid diseases. Chem Soc Rev 2017; 46:6492-6531. [PMID: 28702523 PMCID: PMC5902192 DOI: 10.1039/c7cs00372b] [Citation(s) in RCA: 256] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Neurodegenerative disorders and type 2 diabetes are global epidemics compromising the quality of life of millions worldwide, with profound social and economic implications. Despite the significant differences in pathology - much of which are poorly understood - these diseases are commonly characterized by the presence of cross-β amyloid fibrils as well as the loss of neuronal or pancreatic β-cells. In this review, we document research progress on the molecular and mesoscopic self-assembly of amyloid-beta, alpha synuclein, human islet amyloid polypeptide and prions, the peptides and proteins associated with Alzheimer's, Parkinson's, type 2 diabetes and prion diseases. In addition, we discuss the toxicities of these amyloid proteins based on their self-assembly as well as their interactions with membranes, metal ions, small molecules and engineered nanoparticles. Through this presentation we show the remarkable similarities and differences in the structural transitions of the amyloid proteins through primary and secondary nucleation, the common evolution from disordered monomers to alpha-helices and then to β-sheets when the proteins encounter the cell membrane, and, the consensus (with a few exceptions) that off-pathway oligomers, rather than amyloid fibrils, are the toxic species regardless of the pathogenic protein sequence or physicochemical properties. In addition, we highlight the crucial role of molecular self-assembly in eliciting the biological and pathological consequences of the amyloid proteins within the context of their cellular environments and their spreading between cells and organs. Exploiting such structure-function-toxicity relationship may prove pivotal for the detection and mitigation of amyloid diseases.
Collapse
Affiliation(s)
- Pu Chun Ke
- ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Marc-Antonie Sani
- School of Chemistry, Bio21 Institute, The University of Melbourne, 30 Flemington Rd, Parkville, VIC 3010, Australia
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
| | - Aleksandr Kakinen
- ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Ibrahim Javed
- ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Frances Separovic
- School of Chemistry, Bio21 Institute, The University of Melbourne, 30 Flemington Rd, Parkville, VIC 3010, Australia
| | - Thomas P. Davis
- ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Department of Chemistry, University of Warwick, Gibbet Hill, Coventry, CV4 7AL, United Kingdom
| | - Raffaele Mezzenga
- ETH Zurich, Department of Health Science & Technology, Schmelzbergstrasse 9, LFO, E23, 8092 Zurich, Switzerland
| |
Collapse
|
62
|
Maiti P, Manna J, Dunbar GL. Current understanding of the molecular mechanisms in Parkinson's disease: Targets for potential treatments. Transl Neurodegener 2017; 6:28. [PMID: 29090092 PMCID: PMC5655877 DOI: 10.1186/s40035-017-0099-z] [Citation(s) in RCA: 328] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 10/09/2017] [Indexed: 12/21/2022] Open
Abstract
Gradual degeneration and loss of dopaminergic neurons in the substantia nigra, pars compacta and subsequent reduction of dopamine levels in striatum are associated with motor deficits that characterize Parkinson’s disease (PD). In addition, half of the PD patients also exhibit frontostriatal-mediated executive dysfunction, including deficits in attention, short-term working memory, speed of mental processing, and impulsivity. The most commonly used treatments for PD are only partially or transiently effective and are available or applicable to a minority of patients. Because, these therapies neither restore the lost or degenerated dopaminergic neurons, nor prevent or delay the disease progression, the need for more effective therapeutics is critical. In this review, we provide a comprehensive overview of the current understanding of the molecular signaling pathways involved in PD, particularly within the context of how genetic and environmental factors contribute to the initiation and progression of this disease. The involvement of molecular chaperones, autophagy-lysosomal pathways, and proteasome systems in PD are also highlighted. In addition, emerging therapies, including pharmacological manipulations, surgical procedures, stem cell transplantation, gene therapy, as well as complementary, supportive and rehabilitation therapies to prevent or delay the progression of this complex disease are reviewed.
Collapse
Affiliation(s)
- Panchanan Maiti
- Field Neurosciences Institute Laboratory for Restorative Neurology, Mt. Pleasant, MI 48859 USA.,Program in Neuroscience, Mt. Pleasant, MI 48859 USA.,Department of Psychology, Central Michigan University, Mt. Pleasant, MI 48859 USA.,Field Neurosciences Institute, St. Mary's of Michigan, Saginaw, MI 48604 USA.,Department of Biology, Saginaw Valley State University, Saginaw, MI 48604 USA
| | - Jayeeta Manna
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38105 USA
| | - Gary L Dunbar
- Field Neurosciences Institute Laboratory for Restorative Neurology, Mt. Pleasant, MI 48859 USA.,Program in Neuroscience, Mt. Pleasant, MI 48859 USA.,Department of Psychology, Central Michigan University, Mt. Pleasant, MI 48859 USA.,Field Neurosciences Institute, St. Mary's of Michigan, Saginaw, MI 48604 USA
| |
Collapse
|
63
|
Dhillon JKS, Riffe C, Moore BD, Ran Y, Chakrabarty P, Golde TE, Giasson BI. A novel panel of α-synuclein antibodies reveal distinctive staining profiles in synucleinopathies. PLoS One 2017; 12:e0184731. [PMID: 28910367 PMCID: PMC5599040 DOI: 10.1371/journal.pone.0184731] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 08/24/2017] [Indexed: 12/25/2022] Open
Abstract
Synucleinopathies are a spectrum of neurodegenerative diseases characterized by the intracellular deposition of the protein α-synuclein leading to multiple outcomes, including dementia and Parkinsonism. Recent findings support the notion that across the spectrum of synucleinopathies there exist diverse but specific biochemical modifications and/or structural conformations of α-synuclein, which would give rise to protein strain specific prion-like intercellular transmission, a proposed model that could explain synucleinopathies disease progression. Herein, we characterized a panel of antibodies with epitopes within both the C- and N- termini of α-synuclein. A comprehensive analysis of human pathological tissue and mouse models of synucleinopathy with these antibodies support the notion that α-synuclein exists in distinct modified forms and/or structural variants. Furthermore, these well-characterized and specific tools allow the investigation of biochemical changes associated with α-synuclein inclusion formation. We have identified several antibodies of interest with diverse staining and epitope properties that will prove useful in future investigations of strain specific disease progression and the development of targeted immunotherapeutic approaches to synucleinopathies.
Collapse
Affiliation(s)
- Jess-Karan S. Dhillon
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, College of Medicine University of Florida, Gainesville, Florida, United States of America
| | - Cara Riffe
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, College of Medicine University of Florida, Gainesville, Florida, United States of America
| | - Brenda D. Moore
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, College of Medicine University of Florida, Gainesville, Florida, United States of America
| | - Yong Ran
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, College of Medicine University of Florida, Gainesville, Florida, United States of America
| | - Paramita Chakrabarty
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, College of Medicine University of Florida, Gainesville, Florida, United States of America
| | - Todd E. Golde
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, College of Medicine University of Florida, Gainesville, Florida, United States of America
| | - Benoit I. Giasson
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, College of Medicine University of Florida, Gainesville, Florida, United States of America
- * E-mail:
| |
Collapse
|
64
|
von Euler Chelpin M, Vorup-Jensen T. Targets and Mechanisms in Prevention of Parkinson's Disease through Immunomodulatory Treatments. Scand J Immunol 2017; 85:321-330. [PMID: 28231624 DOI: 10.1111/sji.12542] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 02/18/2017] [Indexed: 01/13/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease in the world; however, there is no cure for it. Current treatments only relieve some of the symptoms, without ceasing the disease, and lose efficacy with prolonged treatment. Considerable evidence shows that persistent inflammatory responses, involving T cell infiltration and glial cell activation, are common characteristics of human patients and play a crucial role in the degeneration of dopaminergic neurons. Therefore, it is important to develop therapeutic strategies that can impede or halt the disease through the modulation of the peripheral immune system by aiming at controlling the existing neuroinflammation. Most of the immunomodulatory therapies designed for the treatment of Parkinson's disease are based on vaccines using AS or antibodies against it; yet, it is of significant interest to explore other formulations that could be used as therapeutic agents. Several vaccination procedures have shown that inducing regulatory T cells in the periphery is protective in PD animal models. In this regard, the formulation glatiramer acetate (Copaxone® ), extensively used for the treatment of multiple sclerosis, could be a suitable candidate due to its capability to increase the number and suppressor capacity of regulatory T cells. In this review, we will present some of the recent immunomodulatory therapies for PD including vaccinations with AS or glatiramoids, or both, as treatments of PD pathology.
Collapse
Affiliation(s)
| | - T Vorup-Jensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
65
|
El-Agnaf O, Overk C, Rockenstein E, Mante M, Florio J, Adame A, Vaikath N, Majbour N, Lee SJ, Kim C, Masliah E, Rissman RA. Differential effects of immunotherapy with antibodies targeting α-synuclein oligomers and fibrils in a transgenic model of synucleinopathy. Neurobiol Dis 2017; 104:85-96. [PMID: 28476636 PMCID: PMC5954414 DOI: 10.1016/j.nbd.2017.05.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/25/2017] [Accepted: 05/01/2017] [Indexed: 11/17/2022] Open
Abstract
Disorders with progressive accumulation of α-synuclein (α-syn) are a common cause of dementia and parkinsonism in the aging population. Accumulation and propagation of α-syn play a role in the pathogenesis of these disorders. Previous studies have shown that immunization with antibodies that recognize C-terminus of α-syn reduces the intra-neuronal accumulation of α-syn and related deficits in transgenic models of synucleinopathy. These studies employed antibodies that recognize epitopes within monomeric and aggregated α-syn that were generated through active immunization or administered via passive immunization. However, it is possible that more specific effects might be achieved with antibodies recognizing selective species of the α-syn aggregates. In this respect we recently developed antibodies that differentially recognized various oligomers (Syn-O1, -O2, and -O4) and fibrilar (Syn-F1 and -F2) forms of α-syn. For this purpose wild-type α-syn transgenic (line 61) mice were immunized with these 5 different antibodies and neuropathologically and biochemically analyzed to determine which was most effective at reducing α-syn accumulation and related deficits. We found that Syn-O1, -O4 and -F1 antibodies were most effective at reducing accumulation of α-syn oligomers in multiple brain regions and at preventing neurodegeneration. Together this study supports the notion that selective antibodies against α-syn might be suitable for development new treatments for synucleinopathies such as PD and DLB.
Collapse
Affiliation(s)
- Omar El-Agnaf
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, PO Box 5825, Doha, Qatar; Life Sciences Division, College of Science and Engineering, Hamad Bin Khalifa University (HBKU), Education City, Qatar Foundation, PO Box 5825, Doha, Qatar
| | - Cassia Overk
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, United States
| | - Edward Rockenstein
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, United States
| | - Michael Mante
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, United States
| | - Jazmin Florio
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, United States
| | - Anthony Adame
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, United States
| | - Nishant Vaikath
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, PO Box 5825, Doha, Qatar
| | - Nour Majbour
- Neurological Disorders Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, PO Box 5825, Doha, Qatar
| | - Seung-Jae Lee
- Department of Biomedical Sciences and Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Changyoun Kim
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, United States
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, United States; Department of Pathology, University of California, San Diego, La Jolla, CA 92093, United States
| | - Robert A Rissman
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, United States; Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, United States.
| |
Collapse
|
66
|
Sklerov M, Kang UJ, Liong C, Clark L, Marder K, Pauciulo M, Nichols WC, Chung WK, Honig LS, Cortes E, Vonsattel JP, Alcalay RN. Frequency of GBA variants in autopsy-proven multiple system atrophy. Mov Disord Clin Pract 2017; 4:574-581. [PMID: 28966932 PMCID: PMC5614491 DOI: 10.1002/mdc3.12481] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 01/24/2017] [Accepted: 02/02/2017] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Multiple system atrophy (MSA) is marked by abnormal inclusions of alpha-synuclein in oligodendrogliocytes. Etiology remains unknown. Variants in the glucocerebrosidase gene have been associated with other synucleinopathies, dementia with Lewy bodies and Parkinson disease. It is unclear whether glucocerebrosidase variants are associated with MSA. OBJECTIVES To analyze the frequency of glucocerebrosidase gene variants among autopsy-proven cases of MSA at a brain bank in New York City. METHODS The glucocerebrosidase gene was fully sequenced in the 17 autopsy-proven MSA cases with extractable DNA at the Columbia University New York Brain Bank from 2002 to 2016. To test if the MSA cases in the brain bank are enriched for GBA variants, we compared the GBA variant frequency in MSA to all brain bank cases with pure Alzheimer's disease (AD) at Columbia University for whom GBA genotype was available (n=82). RESULTS 4/17 (23.5%) MSA cases carried glucocerebrosidase gene variants, including an individual homozygous for N370S, and one each who were heterozygous carriers of N370S, T369M and R496H. Among the comparator cases with pure AD, 3 of the 82 autopsies (3.7%) carried GBA variants (P = 0.0127, Fisher exact test), including one case each of N370S homozygote, and R496H and T369M heterozygous variant. CONCLUSION We found a higher frequency of glucocerebrosidase variants among pathologically diagnosed MSA cases in our brain bank compared to AD autopsies. This study demonstrates the need for further investigation into the role of glucocerebrosidase and lysosomal dysfunction in the etiology of MSA.
Collapse
Affiliation(s)
- Miriam Sklerov
- Department of NeurologyColumbia University Medical CenterNew YorkNew York
| | - Un J. Kang
- Department of NeurologyColumbia University Medical CenterNew YorkNew York
| | - Christopher Liong
- Department of NeurologyColumbia University Medical CenterNew YorkNew York
| | - Lorraine Clark
- Department of Pathology and Cell BiologyNew YorkNew York
| | - Karen Marder
- Department of NeurologyColumbia University Medical CenterNew YorkNew York
| | - Michael Pauciulo
- Department of PediatricsCincinnati Children's Hospital Medical Center and University of Cincinnati College of MedicineCincinnatiOhio
| | - William C. Nichols
- Department of PediatricsCincinnati Children's Hospital Medical Center and University of Cincinnati College of MedicineCincinnatiOhio
| | - Wendy K. Chung
- Department of PediatricsColumbia University Medical Center
| | - Lawrence S. Honig
- Department of NeurologyColumbia University Medical CenterNew YorkNew York
| | - Etty Cortes
- Department of Pathology and Cell BiologyColumbia University Medical Center
| | | | - Roy N. Alcalay
- Department of NeurologyColumbia University Medical CenterNew YorkNew York
| |
Collapse
|
67
|
Arevalo-Villalobos JI, Govea-Alonso DO, Monreal-Escalante E, Zarazúa S, Rosales-Mendoza S. LTB-Syn: a recombinant immunogen for the development of plant-made vaccines against synucleinopathies. PLANTA 2017; 245:1231-1239. [PMID: 28315001 DOI: 10.1007/s00425-017-2675-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 03/08/2017] [Indexed: 06/06/2023]
Abstract
MAIN CONCLUSION A recombinant antigen targeting α-synuclein was produced in the plant cell rendering an immunogenic protein capable to induce humoral responses in mice upon oral administration. Synucleinopathies are neurodegenerative diseases characterized by the abnormal accumulation of α-synuclein (α-Syn, a 140 amino acid protein that normally plays various neurophysiologic roles) aggregates. Parkinson's disease (PD) is the synucleinopathy with the highest epidemiologic impact and although its etiology remains unknown, α-Syn aggregation during disease progression pointed out α-Syn as target in the development of immunotherapies. Herein a chimeric protein, comprising the B subunit of the enterotoxin from enterotoxigenic Escherichia coli and α-Syn epitopes, was expressed in the plant cell having the potential to induce humoral responses following oral immunization. This approach will serve as the basis for the development of oral plant-based vaccines against PD with several potential advantages such as low cost, easy scale-up during production, and easy administration.
Collapse
Affiliation(s)
- Jaime I Arevalo-Villalobos
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí, SLP, 78210, Mexico
| | - Dania O Govea-Alonso
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí, SLP, 78210, Mexico
| | - Elizabeth Monreal-Escalante
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí, SLP, 78210, Mexico
| | - Sergio Zarazúa
- Laboratorio de Neurotoxicología, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí, SLP, 78210, Mexico
| | - Sergio Rosales-Mendoza
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, San Luis Potosí, SLP, 78210, Mexico.
| |
Collapse
|
68
|
Spencer B, Valera E, Rockenstein E, Overk C, Mante M, Adame A, Zago W, Seubert P, Barbour R, Schenk D, Games D, Rissman RA, Masliah E. Anti-α-synuclein immunotherapy reduces α-synuclein propagation in the axon and degeneration in a combined viral vector and transgenic model of synucleinopathy. Acta Neuropathol Commun 2017; 5:7. [PMID: 28086964 PMCID: PMC5237270 DOI: 10.1186/s40478-016-0410-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 12/20/2016] [Indexed: 11/10/2022] Open
Abstract
Neurodegenerative disorders such as Parkinson's Disease (PD), PD dementia (PDD) and Dementia with Lewy bodies (DLB) are characterized by progressive accumulation of α-synuclein (α-syn) in neurons. Recent studies have proposed that neuron-to-neuron propagation of α-syn plays a role in the pathogenesis of these disorders. We have previously shown that antibodies against the C-terminus of α-syn reduce the intra-neuronal accumulation of α-syn and related deficits in transgenic models of synucleinopathy, probably by abrogating the axonal transport and accumulation of α-syn in in vivo models. Here, we assessed the effect of passive immunization against α-syn in a new mouse model of axonal transport and accumulation of α-syn. For these purpose, non-transgenic, α-syn knock-out and mThy1-α-syn tg (line 61) mice received unilateral intra-cerebral injections with a lentiviral (LV)-α-syn vector construct followed by systemic administration of the monoclonal antibody 1H7 (recognizes amino acids 91-99) or control IgG for 3 months. Cerebral α-syn accumulation and axonopathy was assessed by immunohistochemistry and effects on behavior were assessed by Morris water maze. Unilateral LV-α-syn injection resulted in axonal propagation of α-syn in the contra-lateral site with subsequent behavioral deficits and axonal degeneration. Passive immunization with 1H7 antibody reduced the axonal accumulation of α-syn in the contra-lateral side and ameliorated the behavioral deficits. Together this study supports the notion that immunotherapy might improve the deficits in models of synucleinopathy by reducing the axonal propagation and accumulation of α-syn. This represents a potential new mode of action through which α-syn immunization might work.
Collapse
|
69
|
Valera E, Monzio Compagnoni G, Masliah E. Review: Novel treatment strategies targeting alpha-synuclein in multiple system atrophy as a model of synucleinopathy. Neuropathol Appl Neurobiol 2016; 42:95-106. [PMID: 26924723 DOI: 10.1111/nan.12312] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 01/26/2016] [Accepted: 02/01/2016] [Indexed: 12/30/2022]
Abstract
Neurodegenerative disorders with alpha-synuclein (α-syn) accumulation (synucleinopathies) include Parkinson's disease (PD), PD dementia, dementia with Lewy bodies and multiple system atrophy (MSA). Due to the involvement of toxic α-syn aggregates in the molecular origin of these disorders, developing effective therapies targeting α-syn is a priority as a disease-modifying alternative to current symptomatic treatments. Importantly, the clinical and pathological attributes of MSA make this disorder an excellent candidate as a synucleinopathy model for accelerated drug development. Recent therapeutic strategies targeting α-syn in in vivo and in vitro models of MSA, as well as in clinical trials, have been focused on the pathological mechanisms of α-syn synthesis, aggregation, clearance, and/or cell-to-cell propagation of its neurotoxic conformers. Here we summarize the most relevant approaches in this direction, with emphasis on their potential as general synucleinopathy modifiers, and enumerate research areas for potential improvement in MSA drug discovery.
Collapse
Affiliation(s)
- E Valera
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - G Monzio Compagnoni
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - E 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
|
70
|
Prądzińska M, Behrendt I, Astorga-Wells J, Manoilov A, Zubarev RA, Kołodziejczyk AS, Rodziewicz-Motowidło S, Czaplewska P. Application of amide hydrogen/deuterium exchange mass spectrometry for epitope mapping in human cystatin C. Amino Acids 2016; 48:2809-2820. [PMID: 27573935 PMCID: PMC5107209 DOI: 10.1007/s00726-016-2316-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/16/2016] [Indexed: 12/01/2022]
Abstract
Human cystatin C (hCC) is a small cysteine protease inhibitor whose oligomerization by propagated domain swapping is linked to certain neurological disorders. One of the ways to prevent hCC dimerization and fibrillogenesis is to enable its interaction with a proper antibody. Herein, the sites of interaction of hCC with dimer-preventing mouse monoclonal anti-hCC antibodies Cyst28 are studied and compared with the binding sites found for mAb Cyst10 that has almost no effect on hCC dimerization. In addition, hCC epitopes in complexes with native polyclonal antibodies extracted from human serum were studied. The results obtained with hydrogen-deuterium exchange mass spectrometry (HDX MS) were compared with the previous findings made using the excision/extraction MS approach. The main results from the two complementary MS-based approaches are found to be in agreement with each other, with some differences being attributed to the specificity of each method. The findings of the current studies may be important for future design of hCC dimerization inhibitors.
Collapse
Affiliation(s)
- Martyna Prądzińska
- Faculty of Chemistry, Department of Biomedical Chemistry, University of Gdańsk, Wita Stwosza 63, 80-952, Gdańsk, Poland
| | - Izabela Behrendt
- Faculty of Chemistry, Department of Biomedical Chemistry, University of Gdańsk, Wita Stwosza 63, 80-952, Gdańsk, Poland
| | - Juan Astorga-Wells
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, S-171 77, Stockholm, Sweden
- Biomotif AB, 18212, Stockholm, Sweden
| | - Aleksandr Manoilov
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, S-171 77, Stockholm, Sweden
| | - Roman A Zubarev
- Division of Physiological Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, S-171 77, Stockholm, Sweden.
| | - Aleksandra S Kołodziejczyk
- Faculty of Chemistry, Department of Biomedical Chemistry, University of Gdańsk, Wita Stwosza 63, 80-952, Gdańsk, Poland
| | - Sylwia Rodziewicz-Motowidło
- Faculty of Chemistry, Department of Biomedical Chemistry, University of Gdańsk, Wita Stwosza 63, 80-952, Gdańsk, Poland
| | - Paulina Czaplewska
- Intercollegiate Faculty of Biotechnology, University of Gdańsk-Medical University of Gdańsk, Kładki 24, 80-822, Gdańsk, Poland.
| |
Collapse
|
71
|
Oh SH, Kim HN, Park HJ, Shin JY, Kim DY, Lee PH. The Cleavage Effect of Mesenchymal Stem Cell and Its Derived Matrix Metalloproteinase-2 on Extracellular α-Synuclein Aggregates in Parkinsonian Models. Stem Cells Transl Med 2016; 6:949-961. [PMID: 28297586 PMCID: PMC5442774 DOI: 10.5966/sctm.2016-0111] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 08/29/2016] [Indexed: 01/01/2023] Open
Abstract
Ample evidence has suggested that extracellular α‐synuclein aggregates would play key roles in the pathogenesis and progression of Parkinsonian disorders (PDs). In the present study, we investigated whether mesenchymal stem cells (MSCs) and their derived soluble factors could exert neuroprotective effects via proteolysis of extracellular α‐synuclein. When preformed α‐synuclein aggregates were incubated with MSC‐conditioned medium, α‐synuclein aggregates were disassembled, and insoluble and oligomeric forms of α‐synuclein were markedly decreased, thus leading to a significant increase in neuronal viability. In an animal study, MSC or MSC‐conditioned medium treatment decreased the expression of α‐synuclein oligomers and the induction of pathogenic α‐synuclein with an attenuation of apoptotic cell death signaling. Furthermore, we identified that matrix metalloproteinase‐2 (MMP‐2), a soluble factor derived from MSCs, played an important role in the degradation of extracellular α‐synuclein. Our data demonstrated that MSCs and their derived MMP‐2 exert neuroprotective properties through proteolysis of aggregated α‐synuclein in PD‐related microenvironments. Stem Cells Translational Medicine2017;6:949–961
Collapse
Affiliation(s)
- Se Hee Oh
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ha Na Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyun Jung Park
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Jin Young Shin
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Dong Yeol Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Phil Hyu Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Severance Biomedical Science Institute, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
72
|
Arevalo-Villalobos JI, Rosales-Mendoza S, Zarazua S. Immunotherapies for neurodegenerative diseases: current status and potential of plant-made biopharmaceuticals. Expert Rev Vaccines 2016; 16:151-159. [DOI: 10.1080/14760584.2016.1229602] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Jaime I Arevalo-Villalobos
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - Sergio Rosales-Mendoza
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - Sergio Zarazua
- Laboratorio de Neurotoxicología, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| |
Collapse
|
73
|
Spencer B, Williams S, Rockenstein E, Valera E, Xin W, Mante M, Florio J, Adame A, Masliah E, Sierks MR. α-synuclein conformational antibodies fused to penetratin are effective in models of Lewy body disease. Ann Clin Transl Neurol 2016; 3:588-606. [PMID: 27606342 PMCID: PMC4999592 DOI: 10.1002/acn3.321] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/12/2016] [Accepted: 04/29/2016] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Progressive accumulation of α-synuclein (α-syn) has been associated with Parkinson's disease (PD) and Dementia with Lewy body (DLB). The mechanisms through which α-syn leads to neurodegeneration are not completely clear; however, the formation of various oligomeric species have been proposed to play a role. Antibody therapy has shown effectiveness at reducing α-syn accumulation in the central nervous system (CNS); however, most of these studies have been conducted utilizing antibodies that recognize both monomeric and higher molecular weight α-syn. In this context, the main objective of this study was to investigate the efficacy of immunotherapy with single-chain antibodies (scFVs) against specific conformational forms of α-syn fused to a novel brain penetrating sequence. METHOD We screened various scFVs against α-syn expressed from lentiviral vectors by intracerebral injections in an α-syn tg model. The most effective scFVs were fused to the cell-penetrating peptide penetratin to enhance transport across the blood-brain barrier, and lentiviral vectors were constructed and tested for efficacy following systemic delivery intraperitoneal into α-syn tg mice. RESULT Two scFVs (D5 and 10H) selectively targeted different α-syn oligomers and reduced the accumulation of α-syn and ameliorated functional deficits when delivered late in disease development; however, only one of the antibodies (D5) was also effective when delivered early in disease development. These scFVs were also utilized in an enzyme-linked immunosorbent assay (ELISA) assay to monitor the effects of immunotherapy on α-syn oligomers in brain and plasma. INTERPRETATION The design and targeting of antibodies for specific species of α-syn oligomers is crucial for therapeutic immunotherapy and might be of relevance for the treatment of Lewy body disease.
Collapse
Affiliation(s)
- Brian Spencer
- Department of NeuroscienceUniversity of CaliforniaSan DiegoCalifornia
| | | | | | - Elvira Valera
- Department of NeuroscienceUniversity of CaliforniaSan DiegoCalifornia
| | - Wei Xin
- Department of Chemical EngineeringArizona State UniversityTempeArizona
| | - Michael Mante
- Department of NeuroscienceUniversity of CaliforniaSan DiegoCalifornia
| | - Jazmin Florio
- Department of NeuroscienceUniversity of CaliforniaSan DiegoCalifornia
| | - Anthony Adame
- Department of NeuroscienceUniversity of CaliforniaSan DiegoCalifornia
| | - Eliezer Masliah
- Department of NeuroscienceUniversity of CaliforniaSan DiegoCalifornia
- Department of PathologyUniversity of CaliforniaSan DiegoCalifornia
| | - Michael R. Sierks
- Department of Chemical EngineeringArizona State UniversityTempeArizona
| |
Collapse
|
74
|
George S, Brundin P. Immunotherapy in Parkinson's Disease: Micromanaging Alpha-Synuclein Aggregation. JOURNAL OF PARKINSONS DISEASE 2016; 5:413-24. [PMID: 26406122 PMCID: PMC4923719 DOI: 10.3233/jpd-150630] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Currently, several α-synuclein immunotherapies are being tested in experimental Parkinson’s disease models and in clinical trials. Recent research has revealed that α-synuclein is not just an intracellular synaptic protein but also exists extracellularly. Moreover, the transfer of misfolded α-synuclein between cells might be a crucial step in the process leading to a progressive increase in deposition of α-synuclein aggregates throughout the Parkinson’s disease brain. The revelation that α-synuclein is present outside cells has increased the interest in antibody-based therapies and opens up for the notion that microglia might play a key role in retarding Parkinson’s disease progression. The objectives of this review are to describe and contrast the use of active and passive immunotherapy in treating α-synucleinopathies and highlight the likely important role of microglia in clearing misfolded α-synuclein from the extracellular space.
Collapse
Affiliation(s)
| | - Patrik Brundin
- Correspondence to: Patrik Brundin, Van Andel Research Insti-tute, Center for Neurodegenerative Science, 333 Bostwick AvenueNE, Grand Rapids, MI 49503, USA.
Tel.: 616 234 5684; Fax: 616 234 5129.
| |
Collapse
|
75
|
Longitudinal live imaging of retinal α-synuclein::GFP deposits in a transgenic mouse model of Parkinson's Disease/Dementia with Lewy Bodies. Sci Rep 2016; 6:29523. [PMID: 27389831 PMCID: PMC4937425 DOI: 10.1038/srep29523] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 06/17/2016] [Indexed: 01/20/2023] Open
Abstract
Abnormal α-synuclein (α-syn) accumulation in the CNS may underlie neuronal cell and synaptic dysfunction leading to motor and cognitive deficits in synucleinopathies including Parkinson’s disease (PD) and Dementia with Lewy Bodies (DLB). Multiple groups demonstrated α-syn accumulation in CNS accessory structures, including the eyes and olfactory terminals, as well as in peripheral organs of Parkinsonian patients. Retinal imaging studies of mice overexpressing fused α-syn::GFP were conducted to evaluate the presence and progression of retinal pathology in a PD/DLB transgenic mouse model. Bright-field image retinal maps and fluorescent images were acquired at 1-month intervals for 3 months. Retinal imaging revealed the accumulation of GFP-tagged α-syn in retinal ganglion cell layer and in the edges of arterial blood vessels in the transgenic mice. Double labeling studies confirmed that the α-syn::GFP-positive cells were retinal ganglion cells containing α-syn. Accumulation of α-syn persisted in the same cells and increased with age. Accumulation of α-syn::GFP was reduced by immunization with single chain antibodies against α-syn. In conclusion, longitudinal live imaging of the retina in the PDGF-α-syn::GFP mice might represent a useful, non-invasive tool to monitor the fate of α-syn accumulation in the CNS and to evaluate the therapeutic effects of compounds targeting α-syn.
Collapse
|
76
|
Gordon R, Singh N, Lawana V, Ghosh A, Harischandra DS, Jin H, Hogan C, Sarkar S, Rokad D, Panicker N, Anantharam V, Kanthasamy AG, Kanthasamy A. Protein kinase Cδ upregulation in microglia drives neuroinflammatory responses and dopaminergic neurodegeneration in experimental models of Parkinson's disease. Neurobiol Dis 2016; 93:96-114. [PMID: 27151770 DOI: 10.1016/j.nbd.2016.04.008] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 04/22/2016] [Accepted: 04/29/2016] [Indexed: 02/06/2023] Open
Abstract
Chronic microglial activation has been linked to the progressive degeneration of the nigrostriatal dopaminergic neurons evidenced in Parkinson's disease (PD) pathogenesis. The exact etiology of PD remains poorly understood. Although both oxidative stress and neuroinflammation are identified as co-contributors in PD pathogenesis, signaling mechanisms underlying neurodegenerative processes have yet to be defined. Indeed, we recently identified that protein kinase C delta (PKCδ) activation is critical for induction of dopaminergic neuronal loss in response to neurotoxic stressors. However, it remains to be defined whether PKCδ activation contributes to immune signaling events driving microglial neurotoxicity. In the present study, we systematically investigated whether PKCδ contributes to the heightened microglial activation response following exposure to major proinflammatory stressors, including α-synuclein, tumor necrosis factor α (TNFα), and lipopolysaccharide (LPS). We report that exposure to the aforementioned inflammatory stressors dramatically upregulated PKCδ with a concomitant increase in its kinase activity and nuclear translocation in both BV-2 microglial cells and primary microglia. Importantly, we also observed a marked upregulation of PKCδ in the microglia of the ventral midbrain region of PD patients when compared to age-matched controls, suggesting a role for microglial PKCδ in neurodegenerative processes. Further, shRNA-mediated knockdown and genetic ablation of PKCδ in primary microglia blunted the microglial proinflammatory response elicited by the inflammogens, including ROS generation, nitric oxide production, and proinflammatory cytokine and chemokine release. Importantly, we found that PKCδ activated NFκB, a key mediator of inflammatory signaling events, after challenge with inflammatory stressors, and that transactivation of NFκB led to translocation of the p65 subunit to the nucleus, IκBα degradation and phosphorylation of p65 at Ser536. Furthermore, both genetic ablation and siRNA-mediated knockdown of PKCδ attenuated NFκB activation, suggesting that PKCδ regulates NFκB activation subsequent to microglial exposure to inflammatory stimuli. To further investigate the pivotal role of PKCδ in microglial activation in vivo, we utilized pre-clinical models of PD. We found that PKCδ deficiency attenuated the proinflammatory response in the mouse substantia nigra, reduced locomotor deficits and recovered mice from sickness behavior in an LPS-induced neuroinflammation model of PD. Likewise, we found that PKCδ knockout mice treated with MPTP displayed a dampened microglial inflammatory response. Moreover, PKCδ knockout mice exhibited reduced susceptibility to the neurotoxin-induced dopaminergic neurodegeneration and associated motor impairments. Taken together, our studies propose a pivotal role for PKCδ in PD pathology, whereby sustained PKCδ activation drives sustained microglial inflammatory responses and concomitant dopaminergic neurotoxicity consequently leading to neurobehavioral deficits. We conclude that inhibiting PKCδ activation may represent a novel therapeutic strategy in PD treatment.
Collapse
Affiliation(s)
- Richard Gordon
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Neeraj Singh
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Vivek Lawana
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Anamitra Ghosh
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Dilshan S Harischandra
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Huajun Jin
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Colleen Hogan
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Souvarish Sarkar
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Dharmin Rokad
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Nikhil Panicker
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Vellareddy Anantharam
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Anumantha G Kanthasamy
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Arthi Kanthasamy
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
77
|
Ugrumov MV. [Development of preclinical diagnosis and preventive treatment of neurodegenerative diseases]. Zh Nevrol Psikhiatr Im S S Korsakova 2016; 115:4-14. [PMID: 26978045 DOI: 10.17116/jnevro20151151114-14] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Neurodegenerative diseases (NDD) are serious fatal neurological and mental diseases that resulted in disability and fethal outcome. Based on the advances of basic sciences over the last two decades, new knowledge on the risk factors for NDD and molecular mechanisms of the pathogenesis are obtained. It has been shown that the accelerated process of neuronal death which is the main cause of NDD development begins long before the appearance of clinical symptoms. The first symptoms appeared only after the death of most specific regulatory neurons and exhaustion of brain compensatory reserve. Only at that time, one can make the diagnosis and start traditional treatment of patients that accounts for the extremely low efficacy of the latter. Currently, complex preclinical diagnosis based on the identification of relatively specific clinical precursors and peripheral biomarkers has been developing. Development of preclinical diagnosis and preventive treatment is a strategic issue of modern neurology and psychiatry. The resolution of this issue allows to consider NDD as cured, but not fatal, diseases.
Collapse
Affiliation(s)
- M V Ugrumov
- Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
78
|
Valera E, Masliah E. Therapeutic approaches in Parkinson's disease and related disorders. J Neurochem 2016; 139 Suppl 1:346-352. [PMID: 26749150 DOI: 10.1111/jnc.13529] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 01/04/2016] [Accepted: 01/05/2016] [Indexed: 01/13/2023]
Abstract
The lack of effective therapies for neurodegenerative disorders is one of the most relevant challenges of this century, considering that, as the global population ages, the incidence of these type of diseases is quickly on the rise. Among these disorders, synucleinopathies, which are characterized by the abnormal accumulation and spreading of the synaptic protein alpha-synuclein in the brain, already constitute the second leading cause of parkinsonism and dementia in the elderly population. Disorders with alpha-synuclein accumulation include Parkinson's disease, dementia with Lewy bodies and multiple system atrophy. Numerous therapeutic alternatives for synucleinopathies are being tested in pre-clinical models and in the clinic; however, only palliative treatments addressing the dopaminergic deficits are approved to date, and no disease-modifying options are available yet. In this article, we provide a brief overview of therapeutic approaches currently being explored for synucleinopathies, and suggest possible explanations to the clinical trials outcomes. Finally, we propose that a deeper understanding of the pathophysiology of synucleinopathies, together with a combination of therapies tailored to each disease stage, may lead to better therapeutic outcomes in synucleinopathy patients. Synucleinopathies, neurodegenerative disorders characterized by the abnormal accumulation of the protein alpha-synuclein, constitute the second leading cause of parkinsonism and dementia in the elderly population, however, no disease-modifying options are available yet. In this review, we summarize the therapeutic approaches currently being explored for synucleinopathies, suggest possible explanations to the clinical outcomes, and propose areas of further therapeutic improvement. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Elvira Valera
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA. .,Department of Pathology, University of California, San Diego, La Jolla, California, USA.
| |
Collapse
|
79
|
Oh SH, Kim HN, Park HJ, Shin JY, Bae EJ, Sunwoo MK, Lee SJ, Lee PH. Mesenchymal Stem Cells Inhibit Transmission of α-Synuclein by Modulating Clathrin-Mediated Endocytosis in a Parkinsonian Model. Cell Rep 2016; 14:835-849. [DOI: 10.1016/j.celrep.2015.12.075] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 09/11/2015] [Accepted: 12/15/2015] [Indexed: 02/06/2023] Open
|
80
|
Valera E, Spencer B, Masliah E. Immunotherapeutic Approaches Targeting Amyloid-β, α-Synuclein, and Tau for the Treatment of Neurodegenerative Disorders. Neurotherapeutics 2016; 13:179-89. [PMID: 26494242 PMCID: PMC4720672 DOI: 10.1007/s13311-015-0397-z] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Disease-modifying alternatives are sorely needed for the treatment of neurodegenerative disorders, a group of diseases that afflict approximately 50 million Americans annually. Immunotherapy is one of the most developed approaches in this direction. Vaccination against amyloid-β, α-synuclein, or tau has been extensively explored, specially as the discovery that these proteins may propagate cell-to-cell and be accessible to antibodies when embedded into the plasma membrane or in the extracellular space. Likewise, the use of passive immunization approaches with specific antibodies against abnormal conformations of these proteins has also yielded promising results. The clinical development of immunotherapies for Alzheimer’s disease, Parkinson’s disease, frontotemporal dementia, dementia with Lewy bodies, and other neurodegenerative disorders is a field in constant evolution. Results to date suggest that immunotherapy is a promising therapeutic approach for neurodegenerative diseases that progress with the accumulation and prion-like propagation of toxic protein aggregates. Here we provide an overview of the most novel and relevant immunotherapeutic advances targeting amyloid-β in Alzheimer’s disease, α-synuclein in Alzheimer’s disease and Parkinson’s disease, and tau in Alzheimer’s disease and frontotemporal dementia.
Collapse
Affiliation(s)
- Elvira Valera
- grid.266100.30000000121074242Department of Neurosciences, University of California, La Jolla, San Diego, CA 92093 USA
| | - Brian Spencer
- grid.266100.30000000121074242Department of Neurosciences, University of California, La Jolla, San Diego, CA 92093 USA
| | - Eliezer Masliah
- grid.266100.30000000121074242Department of Neurosciences, University of California, La Jolla, San Diego, CA 92093 USA
- grid.266100.30000000121074242Department of Pathology, University of California, La Jolla, San Diego, CA 92093 USA
| |
Collapse
|
81
|
|
82
|
Rockenstein E, Ubhi K, Mante M, Florio J, Adame A, Winter S, Brandstaetter H, Meier D, Masliah E. Neuroprotective effects of Cerebrolysin in triple repeat Tau transgenic model of Pick's disease and fronto-temporal tauopathies. BMC Neurosci 2015; 16:85. [PMID: 26611895 PMCID: PMC4662012 DOI: 10.1186/s12868-015-0218-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 11/13/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Tauopathies are a group of neurodegenerative disorders with accumulation of three-repeat (3R) or four-repeat (4R) Tau. While 3R tau is found in Pick's disease and Alzheimer's disease (AD), 4R tau is more abundant in corticobasal degeneration, progressive supranuclear palsy, and AD. We have previously shown that Cerebrolysin™ (CBL), a neuropeptide mixture with neurotrophic effects, ameliorates the pathology in amyloid precursor protein transgenic (tg) mouse model of AD and 4R tau, however it is unclear if CBL ameliorates the deficits and neuropathology in the mouse model of Pick's disease over expressing 3R tau. RESULTS Mice expressing 3R tau (L266V and G272V mutations) under the mThy-1 promoter were treated with CBL in two separate groups, the first was 3 months old (treated for 3 months, IP) and the second was 6 months old (treated for 3 months, IP) at the start of the treatment. We found that although the levels of total 3R tau were unchanged, CBL reduced the levels of hyper-phosphorylated tau in both groups of mice. This was accompanied by reduced neurodegenerative pathology in the neocortex and hippocampus in both groups and by improvements in the behavioral deficits in the nest-building test and water maze in the 3-6 month group. CONCLUSION Taken together these results support the notion that CBL may be beneficial in other taupathy models by reducing the levels of aberrantly phosphorylated tau.
Collapse
Affiliation(s)
- Edward Rockenstein
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, 92093-0624, USA.
| | - Kiren Ubhi
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, 92093-0624, USA.
| | - Michael Mante
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, 92093-0624, USA.
| | - Jazmin Florio
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, 92093-0624, USA.
| | - Anthony Adame
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, 92093-0624, USA.
| | - Stefan Winter
- Clinical Research and Pharmacology, EVER Neuro Pharma GmbH, Unterach, Austria.
| | - Hemma Brandstaetter
- Clinical Research and Pharmacology, EVER Neuro Pharma GmbH, Unterach, Austria.
| | - Dieter Meier
- Clinical Research and Pharmacology, EVER Neuro Pharma GmbH, Unterach, Austria.
| | - Eliezer Masliah
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, 92093-0624, USA.
- Department of Pathology, University of California, La Jolla, San Diego, CA, USA.
| |
Collapse
|
83
|
Vieira BDM, Radford RA, Chung RS, Guillemin GJ, Pountney DL. Neuroinflammation in Multiple System Atrophy: Response to and Cause of α-Synuclein Aggregation. Front Cell Neurosci 2015; 9:437. [PMID: 26778958 PMCID: PMC4700780 DOI: 10.3389/fncel.2015.00437] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 10/19/2015] [Indexed: 11/13/2022] Open
Abstract
Multiple system atrophy (MSA) is a progressive neurodegenerative disease presenting with combinations of autonomic dysfunction, parkinsonism, cerebellar ataxia and/or pyramidal signs. Oligodendroglial cytoplasmic inclusions (GCIs) rich in α-synuclein (α-syn) constitute the disease hallmark, accompanied by neuronal loss and activation of glial cells which indicate neuroinflammation. Recent studies demonstrate that α-syn may be released from degenerating neurons to mediate formation of abnormal inclusion bodies and to induce neuroinflammation which, interestingly, might also favor the formation of intracellular α-syn aggregates as a consequence of cytokine release and the shift to a pro-inflammatory environment. Here, we critically review the relationships between α-syn and astrocytic and microglial activation in MSA to explore the potential of therapeutics which target neuroinflammation.
Collapse
Affiliation(s)
| | - Rowan A Radford
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| | - Roger S Chung
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| | - Gilles J Guillemin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University Sydney, NSW, Australia
| | - Dean L Pountney
- Menzies Health Institute Queensland, Griffith University Gold Coast, QLD, Australia
| |
Collapse
|
84
|
Labrador‐Garrido A, Cejudo‐Guillén M, Daturpalli S, Leal MM, Klippstein R, De Genst EJ, Villadiego J, Toledo‐Aral JJ, Dobson CM, Jackson SE, Pozo D, Roodveldt C. Chaperome screening leads to identification of Grp94/Gp96 and FKBP4/52 as modulators of the α‐synuclein‐elicited immune response. FASEB J 2015; 30:564-77. [DOI: 10.1096/fj.15-275131] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 09/21/2015] [Indexed: 01/01/2023]
Affiliation(s)
- Adahir Labrador‐Garrido
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER)SevilleSpain
- Department of Medical Biochemistry, Molecular Biology, and ImmunologySchool of MedicineUniversity of SevilleSevilleSpain
| | - Marta Cejudo‐Guillén
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER)SevilleSpain
- Department of Medical Biochemistry, Molecular Biology, and ImmunologySchool of MedicineUniversity of SevilleSevilleSpain
| | - Soumya Daturpalli
- Department of ChemistryUniversity of CambridgeCambridgeUnited Kingdom
| | - María M. Leal
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER)SevilleSpain
| | - Rebecca Klippstein
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER)SevilleSpain
- Department of Medical Biochemistry, Molecular Biology, and ImmunologySchool of MedicineUniversity of SevilleSevilleSpain
| | - Erwin J. De Genst
- Department of ChemistryUniversity of CambridgeCambridgeUnited Kingdom
| | - Javier Villadiego
- Department of Medical Physiology and BiophysicsSchool of MedicineUniversity of SevilleSevilleSpain
- Institute of Biomedicine of Seville (IBiS)University Hospital Virgen del RocioConsejo Superior de Investigaciones Científicas (CSIC)University of SevilleSevilleSpain
- Centers for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED)SevilleSpain
| | - Juan J. Toledo‐Aral
- Department of Medical Physiology and BiophysicsSchool of MedicineUniversity of SevilleSevilleSpain
- Institute of Biomedicine of Seville (IBiS)University Hospital Virgen del RocioConsejo Superior de Investigaciones Científicas (CSIC)University of SevilleSevilleSpain
- Centers for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED)SevilleSpain
| | | | - Sophie E. Jackson
- Department of ChemistryUniversity of CambridgeCambridgeUnited Kingdom
| | - David Pozo
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER)SevilleSpain
- Department of Medical Biochemistry, Molecular Biology, and ImmunologySchool of MedicineUniversity of SevilleSevilleSpain
| | - Cintia Roodveldt
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER)SevilleSpain
| |
Collapse
|
85
|
Valera E, Masliah E. Combination therapies: The next logical Step for the treatment of synucleinopathies? Mov Disord 2015; 31:225-34. [PMID: 26388203 DOI: 10.1002/mds.26428] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 08/11/2015] [Indexed: 01/13/2023] Open
Abstract
Currently there are no disease-modifying alternatives for the treatment of most neurodegenerative disorders. The available therapies for diseases such as Parkinson's disease (PD), PD dementia (PDD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA), in which the protein alpha-synuclein (α-Syn) accumulates within neurons and glial cells with toxic consequences, are focused on managing the disease symptoms. However, using strategic drug combinations and/or multi-target drugs might increase the treatment efficiency when compared with monotherapies. Synucleinopathies are complex disorders that progress through several stages, and toxic α-Syn aggregates exhibit prion-like behavior spreading from cell to cell. Therefore, it follows that these neurodegenerative disorders might require equally complex therapeutic approaches to obtain significant and long-lasting results. Hypothetically, therapies aimed at reducing α-Syn accumulation and cell-to-cell transfer, such as immunotherapy against α-Syn, could be combined with agents that reduce neuroinflammation with potential synergistic outcomes. Here we review the current evidence supporting this type of approach, suggesting that such rational therapy combinations, together with the use of multi-target drugs, may hold promise as the next logical step for the treatment of synucleinopathies.
Collapse
Affiliation(s)
- Elvira Valera
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA.,Department of Pathology, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
86
|
Zhang Q, Kim YC, Narayanan NS. Disease-modifying therapeutic directions for Lewy-Body dementias. Front Neurosci 2015; 9:293. [PMID: 26347604 PMCID: PMC4542461 DOI: 10.3389/fnins.2015.00293] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 08/04/2015] [Indexed: 12/26/2022] Open
Abstract
Dementia with Lewy bodies (DLB) is the second leading cause of dementia following Alzheimer's disease (AD) and accounts for up to 25% of all dementia. DLB is distinct from AD in that it involves extensive neuropsychiatric symptoms as well as motor symptoms, leads to enormous societal costs in terms of direct medical care and is associated with high financial and caregiver costs. Although, there are no disease-modifying therapies for DLB, we review several new therapeutic directions in treating DLB. We discuss progress in strategies to decrease the level of alpha-synuclein, to prevent the cell to cell transmission of misfolded alpha-synuclein, and the potential of brain stimulation in DLB.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Neurology, University of Iowa Iowa City, IA, USA ; Physician Scientist Training Program, University of Iowa Iowa City, IA, USA
| | - Young-Cho Kim
- Department of Neurology, University of Iowa Iowa City, IA, USA
| | - Nandakumar S Narayanan
- Department of Neurology, University of Iowa Iowa City, IA, USA ; Aging Mind and Brain Initiative, Carver College of Medicine, University of Iowa Iowa City, IA, USA
| |
Collapse
|
87
|
Schneeberger A, Tierney L, Mandler M. Active immunization therapies for Parkinson's disease and multiple system atrophy. Mov Disord 2015; 31:214-24. [PMID: 26260853 DOI: 10.1002/mds.26377] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/16/2015] [Accepted: 07/20/2015] [Indexed: 01/06/2023] Open
Abstract
Vaccination is increasingly being investigated as a potential treatment for synucleinopathies, a group of neurodegenerative diseases including Parkinson's disease, multiple system atrophy, and dementia with Lewy bodies associated with α-synuclein pathology. All lack a causal therapy. Development of novel, disease-altering treatment strategies is urgently needed. Vaccination has positioned itself as a prime strategy for addressing these diseases because it is broadly applicable, requires infrequent administration, and maintains low production costs for treating a large population or as a preventive measure. Current evidence points to a causal role of misfolded α-synuclein in the development and progression of synucleinopathies. In the past decade, significant progress in active immunization against α-synuclein has been shown both in preclinical animal models and in early clinical development. In this review, we describe the state-of-the-art in active immunization approaches to synucleinopathies, with a focus on advances in Parkinson's disease (PD) and multiple-system atrophy (MSA). We first review preclinical animal models, highlighting their progress in translation to the clinical setting. We then discuss current clinical applications, stressing different approaches taken to address α-synuclein pathology. Finally, we address challenges, trends, and future perspectives of current vaccination programs.
Collapse
|
88
|
Opportunities for Conformation-Selective Antibodies in Amyloid-Related Diseases. Antibodies (Basel) 2015. [DOI: 10.3390/antib4030170] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
89
|
Lawand NB, Saadé NE, El-Agnaf OM, Safieh-Garabedian B. Targeting α-synuclein as a therapeutic strategy for Parkinson's disease. Expert Opin Ther Targets 2015; 19:1351-60. [PMID: 26135549 DOI: 10.1517/14728222.2015.1062877] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION α-Synuclein, a neuronal protein, plays a central role in the pathophysiology of Parkinson's disease (PD), the second most prevalent neurodegenerative disorder. Cases of PD have increased tremendously over the past decade necessitating the identification of new therapeutic targets to reduce patient morbidity and to improve PD patients' quality of life. AREAS COVERED The purpose of this article is to provide an update on the role of α-synuclein in fibrils formation and review its role as an effective immunotherapeutic target for PD. The rapidly expanding evidence for the contribution of α-synuclein to the pathogenesis of PD led to the development of antibodies against the C terminus of α-synuclein and other molecules involved in the inflammatory signaling pathways that were found to contribute significantly to initiation and progression of the disease. EXPERT OPINION The readers will obtain new insights on the mechanisms by which α-synuclein can trigger the development of PD and other related degenerative disorders along with the potential role of active and passive antibodies targeted against specific form of α-synuclein aggregates to clear neurotoxicity, stop the propagation of the prion-like behavior of these oligomers and reverse neuronal degeneration associated with PD.
Collapse
Affiliation(s)
- Nada B Lawand
- a 1 American University of Beirut, Department of Anatomy, Cell Biology and Physiology Sciences , Beirut, Lebanon
| | - Nayef E Saadé
- a 1 American University of Beirut, Department of Anatomy, Cell Biology and Physiology Sciences , Beirut, Lebanon
| | - Omar M El-Agnaf
- b 2 Hamad Ben Khalifa University, College of Science and Engineering, Education City, Qatar Foundation , Doha, Qatar
| | - Bared Safieh-Garabedian
- c 3 Qatar University, College of Medicine, Department of Biological and Environmental Sciences , Doha, Qatar
| |
Collapse
|
90
|
Mandler M, Valera E, Rockenstein E, Mante M, Weninger H, Patrick C, Adame A, Schmidhuber S, Santic R, Schneeberger A, Schmidt W, Mattner F, Masliah E. Active immunization against alpha-synuclein ameliorates the degenerative pathology and prevents demyelination in a model of multiple system atrophy. Mol Neurodegener 2015; 10:10. [PMID: 25886309 PMCID: PMC4411775 DOI: 10.1186/s13024-015-0008-9] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 03/02/2015] [Indexed: 12/16/2022] Open
Abstract
Background Multiple system atrophy (MSA) is a neurodegenerative disease characterized by parkinsonism, ataxia and dysautonomia. Histopathologically, the hallmark of MSA is the abnormal accumulation of alpha-synuclein (α-syn) within oligodendroglial cells, leading to neuroinflammation, demyelination and neuronal death. Currently, there is no disease-modifying treatment for MSA. In this sense, we have previously shown that next-generation active vaccination technology with short peptides, AFFITOPEs®, was effective in two transgenic models of synucleinopathies at reducing behavioral deficits, α-syn accumulation and inflammation. Results In this manuscript, we used the most effective AFFITOPE® (AFF 1) for immunizing MBP-α-syn transgenic mice, a model of MSA that expresses α-syn in oligodendrocytes. Vaccination with AFF 1 resulted in the production of specific anti-α-syn antibodies that crossed into the central nervous system and recognized α-syn aggregates within glial cells. Active vaccination with AFF 1 resulted in decreased accumulation of α-syn, reduced demyelination in neocortex, striatum and corpus callosum, and reduced neurodegeneration. Clearance of α-syn involved activation of microglia and reduced spreading of α-syn to astroglial cells. Conclusions This study further validates the efficacy of vaccination with AFFITOPEs® for ameliorating the neurodegenerative pathology in synucleinopathies. Electronic supplementary material The online version of this article (doi:10.1186/s13024-015-0008-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Elvira Valera
- Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| | - Edward Rockenstein
- Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| | - Michael Mante
- Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| | | | - Christina Patrick
- Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| | - Anthony Adame
- Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| | | | | | | | | | - Frank Mattner
- AFFiRiS AG, Vienna Biocenter, A-1030, Vienna, Austria.
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA. .,Department of Pathology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| |
Collapse
|
91
|
Abstract
Sporadic or idiopathic Parkinson's disease (PD) is an age-related neurodegenerative disorder of unknown origin that ranks only second behind Alzheimer's disease (AD) in prevalence and its consequent social and economic burden. PD neuropathology is characterized by a selective loss of dopaminergic neurons in the substantia nigra pars compacta; however, more widespread involvement of other CNS structures and peripheral tissues now is widely documented. The onset of molecular and cellular neuropathology of PD likely occurs decades before the onset of the motor symptoms characteristic of PD. The hallmark symptoms of PD, resting tremors, rigidity and postural disabilities, are related to dopamine (DA) deficiency. Current therapies treat these symptoms by replacing or boosting existing DA. All current interventions have limited therapeutic benefit for disease progression because damage likely has progressed over an estimated period of ~5 to 15years to a loss of 60%-80% of the nigral DA neurons, before symptoms emerge. There is no accepted definitive biomarker of PD. An urgent need exists to develop early diagnostic biomarkers for two reasons: (1) to intervene at the onset of disease and (2) to monitor the progress of therapeutic interventions that may slow or stop the course of the disease. In the context of disease development, one of the promises of personalized medicine is the ability to predict, on an individual basis, factors contributing to the susceptibility for the development of a given disease. Recent advances in our understanding of genetic factors underlying or contributing to PD offer the potential for monitoring susceptibility biomarkers that can be used to identify at-risk individuals and possibly prevent the onset of disease through treatment. Finally, the exposome concept is new in the biomarker discovery arena and it is suggested as a way to move forward in identifying biomarkers of neurological diseases. It is a two-stage scheme involving a first stage of exposome-wide association studies (EWAS) to profile omic features in serum to discover molecular biomarkers. The second stage involves application of this knowledge base in follow-up studies. This strategy is unique in that it promotes the use of data-driven (omic) strategies in interrogating diseased and healthy populations and encourages a movement away from using only reductionist strategies to discover biomarkers of exposure and disease. In this short review we will examine 1) advances in our understanding of the molecular mechanisms underlying PD that have led to candidate biomarkers for diagnosis and treatment efficacy and 2) new technologies on the horizon that will lead to novel approaches in biomarker development.
Collapse
Affiliation(s)
- Diane B Miller
- Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, WV 26505.
| | - James P O'Callaghan
- Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, WV 26505.
| |
Collapse
|
92
|
Shahaduzzaman M, Nash K, Hudson C, Sharif M, Grimmig B, Lin X, Bai G, Liu H, Ugen KE, Cao C, Bickford PC. Anti-human α-synuclein N-terminal peptide antibody protects against dopaminergic cell death and ameliorates behavioral deficits in an AAV-α-synuclein rat model of Parkinson's disease. PLoS One 2015; 10:e0116841. [PMID: 25658425 PMCID: PMC4319932 DOI: 10.1371/journal.pone.0116841] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 12/15/2014] [Indexed: 12/18/2022] Open
Abstract
The protein α-synuclein (α-Syn) has a central role in the pathogenesis of Parkinson’s disease (PD) and immunotherapeutic approaches targeting this molecule have shown promising results. In this study, novel antibodies were generated against specific peptides from full length human α-Syn and evaluated for effectiveness in ameliorating α-Syn-induced cell death and behavioral deficits in an AAV-α-Syn expressing rat model of PD. Fisher 344 rats were injected with rAAV vector into the right substantia nigra (SN), while control rats received an AAV vector expressing green fluorescent protein (GFP). Beginning one week after injection of the AAV-α-Syn vectors, rats were treated intraperitoneally with either control IgG or antibodies against the N-terminal (AB1), or central region (AB2) of α-Syn. An unbiased stereological estimation of TH+, NeuN+, and OX6 (MHC-II) immunostaining revealed that the α-Syn peptide antibodies (AB1 and AB2) significantly inhibited α-Syn-induced dopaminergic cell (DA) and NeuN+ cell loss (one-way ANOVA (F (3, 30) = 5.8, p = 0.002 and (F (3, 29) = 7.92, p = 0.002 respectively), as well as decreasing the number of activated microglia in the ipsilateral SN (one-way ANOVA F = 14.09; p = 0.0003). Antibody treated animals also had lower levels of α-Syn in the ipsilateral SN (one-way ANOVA F (7, 37) = 9.786; p = 0.0001) and demonstrated a partial intermediate improvement of the behavioral deficits. Our data suggest that, in particular, an α-Syn peptide antibody against the N-terminal region of the protein can protect against DA neuron loss and, to some extent behavioral deficits. As such, these results may be a potential therapeutic strategy for halting the progression of PD.
Collapse
Affiliation(s)
- Md Shahaduzzaman
- Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, Florida, 33612, United States of America
| | - Kevin Nash
- Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, Florida, 33612, United States of America
- USF-Health Byrd Alzheimer’s Institute University of South Florida, Tampa, Florida, 33612, United States of America
| | - Charles Hudson
- James A. Haley Veterans Affairs Hospital, Research Service, Tampa, Florida, 33612, United States of America
| | - Masroor Sharif
- Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, Florida, 33612, United States of America
| | - Bethany Grimmig
- Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, Florida, 33612, United States of America
| | - Xiaoyang Lin
- USF-Health Byrd Alzheimer’s Institute University of South Florida, Tampa, Florida, 33612, United States of America
| | - Ge Bai
- USF-Health Byrd Alzheimer’s Institute University of South Florida, Tampa, Florida, 33612, United States of America
| | - Hui Liu
- USF-Health Byrd Alzheimer’s Institute University of South Florida, Tampa, Florida, 33612, United States of America
| | - Kenneth E. Ugen
- Dept. of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, Florida, 33612, United States of America
- Center for Molecular Delivery, University of South Florida, Tampa, Florida, 33620, United States of America
| | - Chuanhai Cao
- USF-Health Byrd Alzheimer’s Institute University of South Florida, Tampa, Florida, 33612, United States of America
- Dept. of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, Florida, 33612, United States of America
- * E-mail: (PB); (CC)
| | - Paula C. Bickford
- Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, Florida, 33612, United States of America
- Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, Florida, 33612, United States of America
- James A. Haley Veterans Affairs Hospital, Research Service, Tampa, Florida, 33612, United States of America
- * E-mail: (PB); (CC)
| |
Collapse
|
93
|
Valadas JS, Vos M, Verstreken P. Therapeutic strategies in Parkinson's disease: what we have learned from animal models. Ann N Y Acad Sci 2014; 1338:16-37. [PMID: 25515068 DOI: 10.1111/nyas.12577] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 09/30/2014] [Accepted: 10/13/2014] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD), the second most common neurodegenerative disorder, is characterized by a loss of dopaminergic neurons in the substantia nigra, as well as in other brain areas. The currently available dopamine replacement therapy provides merely symptomatic benefit and is ineffective because habituation and side effects arise relatively quickly. Studying the genetic forms of PD in animal models provides novel insight that allows targeting of specific aspects of this heterogenic disease more specifically. Among others, two important cellular deficits are associated with PD; these deficits relate to (1) synaptic transmission and vesicle trafficking, and (2) mitochondrial function, relating respectively to the dominant and recessive mutations in PD-causing genes. With increased knowledge of PD, the possibility of identifying an efficient, long-lasting treatment is becoming more conceivable, but this can only be done with an increased knowledge of the specific affected cellular mechanisms. This review discusses how discoveries in animal models of PD have clarified the therapeutic potential of pathways disrupted in PD, with a specific focus on synaptic transmission, vesicle trafficking, and mitochondrial function.
Collapse
Affiliation(s)
- Jorge S Valadas
- VIB Center for the Biology of Disease; Department of Human Genetics, Leuven Research Institute for Neuroscience and Disease (LIND), KU Leuven, Leuven, Belgium
| | | | | |
Collapse
|
94
|
Labrador-Garrido A, Cejudo-Guillén M, Klippstein R, De Genst EJ, Tomas-Gallardo L, Leal MM, Villadiego J, Toledo-Aral JJ, Dobson CM, Pozo D, Roodveldt C. Chaperoned amyloid proteins for immune manipulation: α-Synuclein/Hsp70 shifts immunity toward a modulatory phenotype. IMMUNITY INFLAMMATION AND DISEASE 2014; 2:226-38. [PMID: 25866630 PMCID: PMC4386917 DOI: 10.1002/iid3.39] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 09/23/2014] [Accepted: 10/20/2014] [Indexed: 01/02/2023]
Abstract
α-Synuclein (αSyn) is a 140-residue amyloid-forming protein whose aggregation is linked to Parkinson's disease (PD). It has also been found to play a critical role in the immune imbalance that accompanies disease progression, a characteristic that has prompted the search for an effective αSyn-based immunotherapy. In this study, we have simultaneously exploited two important features of certain heat-shock proteins (HSPs): their classical “chaperone” activities and their recently discovered and diverse “immunoactive” properties. In particular, we have explored the immune response elicited by immunization of C57BL/6 mice with an αSyn/Hsp70 protein combination in the absence of added adjuvant. Our results show differential effects for mice immunized with the αSyn/Hsp70 complex, including a restrained αSyn-specific (IgM and IgG) humoral response as well as minimized alterations in the Treg (CD4+CD25+Foxp3+) and Teff (CD4+Foxp3−) cell populations, as opposed to significant changes in mice immunized with αSyn and Hsp70 alone. Furthermore, in vitro-stimulated splenocytes from immunized mice showed the lowest relative response against αSyn challenge for the “αSyn/Hsp70” experimental group as measured by IFN-γ and IL-17 secretion, and higher IL-10 levels when stimulated with LPS. Finally, serum levels of Th1-cytokine IFN-γ and immunomodulatory IL-10 indicated a unique shift toward an immunomodulatory/immunoprotective phenotype in mice immunized with the αSyn/Hsp70 complex. Overall, we propose the use of functional “HSP-chaperoned amyloid/aggregating proteins” generated with appropriate HSP-substrate protein combinations, such as the αSyn/Hsp70 complex, as a novel strategy for immune-based intervention against synucleinopathies and other amyloid or “misfolding” neurodegenerative disorders.
Collapse
Affiliation(s)
- Adahir Labrador-Garrido
- CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine Seville, Spain ; Department of Medical Biochemistry Molecular Biology and Immunology School of Medicine, University of Seville Spain
| | - Marta Cejudo-Guillén
- CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine Seville, Spain ; Department of Medical Biochemistry Molecular Biology and Immunology School of Medicine, University of Seville Spain
| | - Rebecca Klippstein
- CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine Seville, Spain ; Department of Medical Biochemistry Molecular Biology and Immunology School of Medicine, University of Seville Spain
| | | | | | - María M Leal
- CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine Seville, Spain
| | - Javier Villadiego
- IBiS Institute of Biomedicine of Seville, University Hospital Virgen del Rocío-CSIC-University of Seville Spain ; Department of Medical Physiology and Biophysics School of Medicine, University of Seville Spain ; CIBERNED, Centers for Networked Biomedical Research in Neurodegenerative Diseases Spain
| | - Juan J Toledo-Aral
- IBiS Institute of Biomedicine of Seville, University Hospital Virgen del Rocío-CSIC-University of Seville Spain ; Department of Medical Physiology and Biophysics School of Medicine, University of Seville Spain ; CIBERNED, Centers for Networked Biomedical Research in Neurodegenerative Diseases Spain
| | | | - David Pozo
- CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine Seville, Spain ; Department of Medical Biochemistry Molecular Biology and Immunology School of Medicine, University of Seville Spain
| | - Cintia Roodveldt
- CABIMER, Andalusian Center for Molecular Biology and Regenerative Medicine Seville, Spain
| |
Collapse
|
95
|
Kim WS, Kågedal K, Halliday GM. Alpha-synuclein biology in Lewy body diseases. ALZHEIMERS RESEARCH & THERAPY 2014; 6:73. [PMID: 25580161 PMCID: PMC4288216 DOI: 10.1186/s13195-014-0073-2] [Citation(s) in RCA: 251] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
α-Synuclein is an abundantly expressed neuronal protein that is at the center of
focus in understanding a group of neurodegenerative disorders called
α-synucleinopathies, which are characterized by the presence of aggregated
α-synuclein intracellularly. Primary α-synucleinopathies include
Parkinson’s disease (PD), dementia with Lewy bodies and multiple system
atrophy, with α-synuclein also found secondarily in a number of other diseases,
including Alzheimer’s disease. Understanding how α-synuclein aggregates
form in these different disorders is important for the understanding of its
pathogenesis in Lewy body diseases. PD is the most prevalent of the
α-synucleinopathies and much of the initial research on α-synuclein Lewy
body pathology was based on PD but is also relevant to Lewy bodies in other diseases
(dementia with Lewy bodies and Alzheimer’s disease). Polymorphism and mutation
studies of SNCA, the gene that encodes α-synuclein, provide much
evidence for a causal link between α-synuclein and PD. Among the primary
α-synucleinopathies, multiple system atrophy is unique in that α-synuclein
deposition occurs in oligodendrocytes rather than neurons. It is unclear whether
α-synuclein originates from oligodendrocytes or whether it is transmitted
somehow from neurons. α-Synuclein exists as a natively unfolded monomer in the
cytosol, but in the presence of lipid membranes it is thought to undergo a
conformational change to a folded α-helical secondary structure that is prone to
forming dimers and oligomers. Posttranslational modification of α-synuclein,
such as phosphorylation, ubiquitination and nitration, has been widely implicated in
α-synuclein aggregation process and neurotoxicity. Recent studies using animal
and cell models, as well as autopsy studies of patients with neuron transplants,
provided compelling evidence for prion-like propagation of α-synuclein. This
observation has implications for therapeutic strategies, and much recent effort is
focused on developing antibodies that target extracellular α-synuclein.
Collapse
Affiliation(s)
- Woojin Scott Kim
- Neuroscience Research Australia, Barker Street, Randwick 2031, NSW, Australia ; School of Medical Sciences, University of New South Wales, Sydney 2052, NSW, Australia
| | - Katarina Kågedal
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, SE-581 85, Sweden
| | - Glenda M Halliday
- Neuroscience Research Australia, Barker Street, Randwick 2031, NSW, Australia ; School of Medical Sciences, University of New South Wales, Sydney 2052, NSW, Australia
| |
Collapse
|
96
|
Gauhar A, Shaykhalishahi H, Gremer L, Mirecka EA, Hoyer W. Impact of subunit linkages in an engineered homodimeric binding protein to α-synuclein. Protein Eng Des Sel 2014; 27:473-9. [PMID: 25332193 DOI: 10.1093/protein/gzu047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Aggregation of the protein α-synuclein (α-syn) has been implicated in Parkinson's disease and other neurodegenerative disorders, collectively referred to as synucleinopathies. The β-wrapin AS69 is a small engineered binding protein to α-syn that stabilizes a β-hairpin conformation of monomeric α-syn and inhibits α-syn aggregation at substoichiometric concentrations. AS69 is a homodimer whose subunits are linked via a disulfide bridge between their single cysteine residues, Cys-28. Here we show that expression of a functional dimer as a single polypeptide chain is achievable by head-to-tail linkage of AS69 subunits. Choice of a suitable linker is essential for construction of head-to-tail dimers that exhibit undiminished α-syn affinity compared with the solely disulfide-linked dimer. We characterize AS69-GS3, a head-to-tail dimer with a glycine-serine-rich linker, under oxidized and reduced conditions in order to evaluate the impact of the Cys28-disulfide bond on structure, stability and α-syn binding. Formation of the disulfide bond causes compaction of AS69-GS3, increases its thermostability, and is a prerequisite for high-affinity binding to α-syn. Comparison of AS69-GS3 and AS69 demonstrates that head-to-tail linkage promotes α-syn binding by affording accelerated disulfide bond formation.
Collapse
Affiliation(s)
- Aziz Gauhar
- Institute of Physical Biology, Heinrich-Heine-Universität, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Hamed Shaykhalishahi
- Institute of Physical Biology, Heinrich-Heine-Universität, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Lothar Gremer
- Institute of Physical Biology, Heinrich-Heine-Universität, Universitätsstraße 1, 40225 Düsseldorf, Germany Institute of Structural Biochemistry (ICS-6), Research Centre Jülich, 52425 Jülich, Germany
| | - Ewa A Mirecka
- Institute of Physical Biology, Heinrich-Heine-Universität, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Wolfgang Hoyer
- Institute of Physical Biology, Heinrich-Heine-Universität, Universitätsstraße 1, 40225 Düsseldorf, Germany Institute of Structural Biochemistry (ICS-6), Research Centre Jülich, 52425 Jülich, Germany
| |
Collapse
|
97
|
Spencer B, Emadi S, Desplats P, Eleuteri S, Michael S, Kosberg K, Shen J, Rockenstein E, Patrick C, Adame A, Gonzalez T, Sierks M, Masliah E. ESCRT-mediated uptake and degradation of brain-targeted α-synuclein single chain antibody attenuates neuronal degeneration in vivo. Mol Ther 2014; 22:1753-1767. [PMID: 25008355 PMCID: PMC4428402 DOI: 10.1038/mt.2014.129] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 07/01/2014] [Indexed: 12/22/2022] Open
Abstract
Parkinson's disease and dementia with Lewy bodies are neurodegenerative disorders characterized by accumulation of α-synuclein (α-syn). Recently, single-chain fragment variables (scFVs) have been developed against individual conformational species of α-syn. Unlike more traditional monoclonal antibodies, these scFVs will not activate or be endocytosed by Fc receptors. For this study, we investigated an scFV directed against oligomeric α-syn fused to the LDL receptor-binding domain from apolipoprotein B (apoB). The modified scFV showed enhanced brain penetration and was imported into neuronal cells through the endosomal sorting complex required for transport (ESCRT) pathway, leading to lysosomal degradation of α-syn aggregates. Further analysis showed that the scFV was effective at ameliorating neurodegenerative pathology and behavioral deficits observed in the mouse model of dementia with Lewy bodies/Parkinson's disease. Thus, the apoB modification had the effect of both increasing accumulation of the scFV in the brain and directing scFV/α-syn complexes for degradation through the ESCRT pathway, leading to improved therapeutic potential of immunotherapy.
Collapse
Affiliation(s)
| | - Sharareh Emadi
- Department of Chemical Engineering, Arizona State University, Tempe, Arizona, USA
| | - Paula Desplats
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Simona Eleuteri
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Sarah Michael
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Kori Kosberg
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Jay Shen
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Edward Rockenstein
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Christina Patrick
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Anthony Adame
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Tania Gonzalez
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Michael Sierks
- Department of Chemical Engineering, Arizona State University, Tempe, Arizona, USA
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA; Department of Pathology, University of California, San Diego, La Jolla, California, USA.
| |
Collapse
|
98
|
Reducing C-terminal-truncated alpha-synuclein by immunotherapy attenuates neurodegeneration and propagation in Parkinson's disease-like models. J Neurosci 2014; 34:9441-54. [PMID: 25009275 DOI: 10.1523/jneurosci.5314-13.2014] [Citation(s) in RCA: 248] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are common neurodegenerative disorders of the aging population, characterized by progressive and abnormal accumulation of α-synuclein (α-syn). Recent studies have shown that C-terminus (CT) truncation and propagation of α-syn play a role in the pathogenesis of PD/DLB. Therefore, we explored the effect of passive immunization against the CT of α-syn in the mThy1-α-syn transgenic (tg) mouse model, which resembles the striato-nigral and motor deficits of PD. Mice were immunized with the new monoclonal antibodies 1H7, 5C1, or 5D12, all directed against the CT of α-syn. CT α-syn antibodies attenuated synaptic and axonal pathology, reduced the accumulation of CT-truncated α-syn (CT-α-syn) in axons, rescued the loss of tyrosine hydroxylase fibers in striatum, and improved motor and memory deficits. Among them, 1H7 and 5C1 were most effective at decreasing levels of CT-α-syn and higher-molecular-weight aggregates. Furthermore, in vitro studies showed that preincubation of recombinant α-syn with 1H7 and 5C1 prevented CT cleavage of α-syn. In a cell-based system, CT antibodies reduced cell-to-cell propagation of full-length α-syn, but not of the CT-α-syn that lacked the 118-126 aa recognition site needed for antibody binding. Furthermore, the results obtained after lentiviral expression of α-syn suggest that antibodies might be blocking the extracellular truncation of α-syn by calpain-1. Together, these results demonstrate that antibodies against the CT of α-syn reduce levels of CT-truncated fragments of the protein and its propagation, thus ameliorating PD-like pathology and improving behavioral and motor functions in a mouse model of this disease.
Collapse
|
99
|
De Genst E, Messer A, Dobson CM. Antibodies and protein misfolding: From structural research tools to therapeutic strategies. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2014; 1844:1907-1919. [PMID: 25194824 DOI: 10.1016/j.bbapap.2014.08.016] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 08/21/2014] [Accepted: 08/27/2014] [Indexed: 01/24/2023]
Abstract
Protein misfolding disorders, including the neurodegenerative conditions Alzheimer's disease (AD) and Parkinson's disease (PD) represent one of the major medical challenges or our time. The underlying molecular mechanisms that govern protein misfolding and its links with disease are very complex processes, involving the formation of transiently populated but highly toxic molecular species within the crowded environment of the cell and tissue. Nevertheless, much progress has been made in understanding these events in recent years through innovative experiments and therapeutic strategies, and in this review we present an overview of the key roles of antibodies and antibody fragments in these endeavors. We discuss in particular how these species are being used in combination with a variety of powerful biochemical and biophysical methodologies, including a range of spectroscopic and microscopic techniques applied not just in vitro but also in situ and in vivo, both to gain a better understanding of the mechanistic nature of protein misfolding and aggregation and also to design novel therapeutic strategies to combat the family of diseases with which they are associated. This article is part of a Special Issue entitled: Recent advances in molecular engineering of antibody.
Collapse
Affiliation(s)
- Erwin De Genst
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| | - Anne Messer
- Neural Stem Cell Institute, Regenerative Research Foundation, Rensselaer, NY 12144, USA; Department of Biomedical Sciences, University at Albany, Albany, NY 12208, USA
| | - Christopher M Dobson
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| |
Collapse
|
100
|
Abstract
Parkinson’s disease (PD) is a progressively debilitating neurodegenerative syndrome. Although best described as a movement disorder, the condition has prominent autonomic, cognitive, psychiatric, sensory and sleep components. Striatal dopaminergic innervation and nigral neurons are progressively lost, with associated Lewy pathology readily apparent on autopsy. Nevertheless, knowledge of the molecular events leading to this pathophysiology is limited. Current therapies offer symptomatic benefit but they fail to slow progression and patients continue to deteriorate. Recent discoveries in sporadic, Mendelian and more complex forms of parkinsonism provide novel insight into disease etiology; 28 genes, including those encoding alpha-synuclein (SNCA), leucine-rich repeat kinase 2 (LRRK2) and microtubule-associated protein tau (MAPT), have been linked and/or associated with PD. A consensus regarding the affected biological pathways and molecular processes has also started to emerge. In early-onset and more a typical PD, deficits in mitophagy pathways and lysosomal function appear to be prominent. By contrast, in more typical late-onset PD, chronic, albeit subtle, dysfunction in synaptic transmission, early endosomal trafficking and receptor recycling, as well as chaperone-mediated autophagy, provide a unifying synthesis of the molecular pathways involved. Disease-modification (neuroprotection) is no longer such an elusive goal given the unparalleled opportunity for diagnosis, translational neuroscience and therapeutic development provided by genetic discovery.
Collapse
Affiliation(s)
- Michelle K Lin
- Djavad Mowafaghian Centre for Brain Health, Centre for Applied Neurogenetics, Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Matthew J Farrer
- Djavad Mowafaghian Centre for Brain Health, Centre for Applied Neurogenetics, Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|