301
|
Calcium Channel Blockers, Progression to Dementia, and Effects on Amyloid Beta Peptide Production. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015. [PMID: 26221415 PMCID: PMC4499419 DOI: 10.1155/2015/787805] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Previous epidemiologic studies suggest that antihypertensive drugs may be protective against cognitive decline. To determine if subjects enrolled in the University of Kentucky longitudinal aging study who used antihypertensive drugs showed diminished progression to dementia, we used a 3-parameter logistic regression model to compare the rate of progression to dementia for subjects who used any of the five common categories of antihypertensive drugs to those with similar demographic characteristics but who did not use antihypertensives. Regression modeling showed that subjects who used calcium channel blockers (CCBs) but not the other classes of antihypertensives showed a significant decrease in the rate of progression to dementia. Significantly, use of CCBs ameliorated the negative effects of the presence of APOE-4 alleles on cognitive decline. To determine if CCBs could minimize amyloid beta peptide (Aβ1–42) production, H4 neuroglioma cultures transfected to overexpress APP were treated with various CCBs and Aβ1–42 levels and levels of proteins involved in Aβ production were quantified. Results show that treatment with nifedipine led to a significant decrease in levels of Aβ1–42, with no significant decrease in cell viability. Collectively, these data suggest that use of CCBs significantly diminishes the rate of progression to dementia and may minimize formation of Aβ1–42.
Collapse
|
302
|
The Inflammatory Form of Cerebral Amyloid Angiopathy or “Cerebral Amyloid Angiopathy-Related Inflammation” (CAARI). Curr Neurol Neurosci Rep 2015; 15:54. [DOI: 10.1007/s11910-015-0572-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
303
|
Abstract
Alzheimer's disease (AD) is one of the most debilitating neurodegenerative diseases and is predicted to affect 1 in 85 people by 2050. Despite much effort to discover a therapeutic strategy to prevent progression or to cure AD, to date no effective disease-modifying agent is available that can prevent, halt, or reverse the cognitive and functional decline of patients with AD. Several underlying etiologies to this failure are proposed. First, accumulating evidence from past trials suggests a preventive as opposed to therapeutic paradigm, and the precise temporal and mechanistic relationship of β-amyloid (Aβ) and tau protein should be elucidated to confirm this hypothesis. Second, we are in urgent need of revised diagnostic criteria to support future trials. Third, various technical and methodological improvements are required, based on the lessons learned from previous failed trials.
Collapse
Affiliation(s)
- Andreas Soejitno
- Department of General Medicine, National Hospital, Jl. Boulevard Famili Selatan Kav.1, Graha Famili, Surabaya, 60228, Indonesia,
| | | | | |
Collapse
|
304
|
Herrup K. The case for rejecting the amyloid cascade hypothesis. Nat Neurosci 2015; 18:794-9. [PMID: 26007212 DOI: 10.1038/nn.4017] [Citation(s) in RCA: 535] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 04/09/2015] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a biologically complex neurodegenerative dementia. Nearly 20 years ago, with the combination of observations from biochemistry, neuropathology and genetics, a compelling hypothesis known as the amyloid cascade hypothesis was formulated. The core of this hypothesis is that it is pathological accumulations of amyloid-β, a peptide fragment of a membrane protein called amyloid precursor protein, that act as the root cause of AD and initiate its pathogenesis. Yet, with the passage of time, growing amounts of data have accumulated that are inconsistent with the basically linear structure of this hypothesis. And while there is fear in the field over the consequences of rejecting it outright, clinging to an inaccurate disease model is the option we should fear most. This Perspective explores the proposition that we are over-reliant on amyloid to define and diagnose AD and that the time has come to face our fears and reject the amyloid cascade hypothesis.
Collapse
Affiliation(s)
- Karl Herrup
- 1] Division of Life Science, Hong Kong University of Science and Technology, Kowloon, Hong Kong. [2] State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Kowloon, Hong Kong
| |
Collapse
|
305
|
Koriyama H, Nakagami H, Nakagami F, Osako MK, Kyutoku M, Shimamura M, Kurinami H, Katsuya T, Rakugi H, Morishita R. Long-Term Reduction of High Blood Pressure by Angiotensin II DNA Vaccine in Spontaneously Hypertensive Rats. Hypertension 2015; 66:167-74. [PMID: 26015450 DOI: 10.1161/hypertensionaha.114.04534] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 04/11/2015] [Indexed: 12/14/2022]
Abstract
Recent research on vaccination has extended its scope from infectious diseases to chronic diseases, including Alzheimer disease, dyslipidemia, and hypertension. The aim of this study was to design DNA vaccines for high blood pressure and eventually develop human vaccine therapy to treat hypertension. Plasmid vector encoding hepatitis B core-angiotensin II (Ang II) fusion protein was injected into spontaneously hypertensive rats using needleless injection system. Anti-Ang II antibody was successfully produced in hepatitis B core-Ang II group, and antibody response against Ang II was sustained for at least 6 months. Systolic blood pressure was consistently lower in hepatitis B core-Ang II group after immunization, whereas blood pressure reduction was continued for at least 6 months. Perivascular fibrosis in heart tissue was also significantly decreased in hepatitis B core-Ang II group. Survival rate was significantly improved in hepatitis B core-Ang II group. This study demonstrated that Ang II DNA vaccine to spontaneously hypertensive rats significantly lowered high blood pressure for at least 6 months. In addition, Ang II DNA vaccines induced an adequate humoral immune response while avoiding the activation of self-reactive T cells, assessed by ELISPOT assay. Future development of DNA vaccine to treat hypertension may provide a new therapeutic option to treat hypertension.
Collapse
Affiliation(s)
- Hiroshi Koriyama
- From the Division of Vascular Medicine and Epigenetics, Osaka University United Graduate School of Child Development, Suita, Osaka, Japan (H.K., H.N., M.K.O., M.S., H.K.); Departments of Clinical Gene Therapy (F.N., M.K., T.K., R.M.) and Geriatric Medicine and Nephrology (F.N., H.R.), Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hironori Nakagami
- From the Division of Vascular Medicine and Epigenetics, Osaka University United Graduate School of Child Development, Suita, Osaka, Japan (H.K., H.N., M.K.O., M.S., H.K.); Departments of Clinical Gene Therapy (F.N., M.K., T.K., R.M.) and Geriatric Medicine and Nephrology (F.N., H.R.), Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Futoshi Nakagami
- From the Division of Vascular Medicine and Epigenetics, Osaka University United Graduate School of Child Development, Suita, Osaka, Japan (H.K., H.N., M.K.O., M.S., H.K.); Departments of Clinical Gene Therapy (F.N., M.K., T.K., R.M.) and Geriatric Medicine and Nephrology (F.N., H.R.), Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Mariana Kiomy Osako
- From the Division of Vascular Medicine and Epigenetics, Osaka University United Graduate School of Child Development, Suita, Osaka, Japan (H.K., H.N., M.K.O., M.S., H.K.); Departments of Clinical Gene Therapy (F.N., M.K., T.K., R.M.) and Geriatric Medicine and Nephrology (F.N., H.R.), Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Mariko Kyutoku
- From the Division of Vascular Medicine and Epigenetics, Osaka University United Graduate School of Child Development, Suita, Osaka, Japan (H.K., H.N., M.K.O., M.S., H.K.); Departments of Clinical Gene Therapy (F.N., M.K., T.K., R.M.) and Geriatric Medicine and Nephrology (F.N., H.R.), Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Munehisa Shimamura
- From the Division of Vascular Medicine and Epigenetics, Osaka University United Graduate School of Child Development, Suita, Osaka, Japan (H.K., H.N., M.K.O., M.S., H.K.); Departments of Clinical Gene Therapy (F.N., M.K., T.K., R.M.) and Geriatric Medicine and Nephrology (F.N., H.R.), Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hitomi Kurinami
- From the Division of Vascular Medicine and Epigenetics, Osaka University United Graduate School of Child Development, Suita, Osaka, Japan (H.K., H.N., M.K.O., M.S., H.K.); Departments of Clinical Gene Therapy (F.N., M.K., T.K., R.M.) and Geriatric Medicine and Nephrology (F.N., H.R.), Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tomohiro Katsuya
- From the Division of Vascular Medicine and Epigenetics, Osaka University United Graduate School of Child Development, Suita, Osaka, Japan (H.K., H.N., M.K.O., M.S., H.K.); Departments of Clinical Gene Therapy (F.N., M.K., T.K., R.M.) and Geriatric Medicine and Nephrology (F.N., H.R.), Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hiromi Rakugi
- From the Division of Vascular Medicine and Epigenetics, Osaka University United Graduate School of Child Development, Suita, Osaka, Japan (H.K., H.N., M.K.O., M.S., H.K.); Departments of Clinical Gene Therapy (F.N., M.K., T.K., R.M.) and Geriatric Medicine and Nephrology (F.N., H.R.), Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Ryuichi Morishita
- From the Division of Vascular Medicine and Epigenetics, Osaka University United Graduate School of Child Development, Suita, Osaka, Japan (H.K., H.N., M.K.O., M.S., H.K.); Departments of Clinical Gene Therapy (F.N., M.K., T.K., R.M.) and Geriatric Medicine and Nephrology (F.N., H.R.), Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| |
Collapse
|
306
|
Abstract
Virus-like particles (VLPs) are an effective means of establishing both prophylactic and therapeutic immunity against their source virus or heterologous antigens. The particulate nature and repetitive structure of VLPs makes them ideal for stimulating potent immune responses. Epitopes delivered by VLPs can be presented on MHC-II for stimulation of a humoral immune response, or cross-presented onto MHC-I leading to cell-mediated immunity. VLPs as particulate subunit vaccine carriers are showing promise in preclinical and clinical trials for the treatment of many conditions including cancer, autoimmunity, allergies and addiction. Supporting the delivery of almost any form of antigenic material, VLPs are ideal candidate vectors for development of future vaccines.
Collapse
|
307
|
Hallé M, Tribout-Jover P, Lanteigne AM, Boulais J, St-Jean JR, Jodoin R, Girouard MP, Constantin F, Migneault A, Renaud F, Didierlaurent AM, Mallett CP, Burkhart D, Pilorget A, Palmantier R, Larocque D. Methods to monitor monocytes-mediated amyloid-beta uptake and phagocytosis in the context of adjuvanted immunotherapies. J Immunol Methods 2015; 424:64-79. [PMID: 26002154 DOI: 10.1016/j.jim.2015.05.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 04/11/2015] [Accepted: 05/06/2015] [Indexed: 12/14/2022]
Abstract
Antibody-mediated capture of amyloid-beta (Aβ) in peripheral blood was identified as an attractive strategy to eliminate cerebral toxic amyloid in Alzheimer's disease (AD) patients and murine models. Alternatively, defective capacity of peripheral monocytes to engulf Aβ was reported in individuals with AD. In this report, we developed different approaches to investigate cellular uptake and phagocytosis of Aβ, and to examine how two immunological devices--an immunostimulatory Adjuvant System and different amyloid specific antibodies--may affect these biological events. Between one and thirteen months of age, APPswe X PS1.M146V (TASTPM) AD model mice had decreasing concentrations of Aβ in their plasma. In contrast, the proportion of blood monocytes containing Aβ tended to increase with age. Importantly, the TLR-agonist containing Adjuvant System AS01B primed monocytes to promote de novo Aβ uptake capacity, particularly in the presence of anti-Aβ antibodies. Biochemical experiments demonstrated that cells achieved Aβ uptake and internalization followed by Aβ degradation via mechanisms that required effective actin polymerization and proteolytic enzymes such as insulin-degrading enzyme. We further demonstrated that both Aβ-specific monoclonal antibodies and plasma from Aβ-immunized mice enhanced the phagocytosis of 1 μm Aβ-coated particles. Together, our data highlight a new biomarker testing to follow amyloid clearance within the blood and a mechanism of Aβ uptake by peripheral monocytes in the context of active or passive immunization, and emphasize on novel approaches to investigate this phenomenon.
Collapse
Affiliation(s)
- Maxime Hallé
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8; Neuroscience Laboratory, 2705, Boulevard Laurier, T-2-50, Department of Molecular Medicine, Centre Hospitalier Universitaire de Québec Research Center, Université Laval, Quebec City, Quebec, Canada, G1V 4G2
| | | | | | - Jonathan Boulais
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8
| | - Julien R St-Jean
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8
| | - Rachel Jodoin
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8
| | | | - Florin Constantin
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8
| | - Annik Migneault
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8
| | - Frédéric Renaud
- GSK Vaccines, Rue de l'Institut 89, B-1330 Rixensart, Belgium
| | | | - Corey P Mallett
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8
| | - David Burkhart
- GSK Vaccines, 553 Old Corvallis Road, Hamilton, MT 59840, USA
| | - Anthony Pilorget
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8
| | - Rémi Palmantier
- GSK Vaccines, Rue de l'Institut 89, B-1330 Rixensart, Belgium
| | - Daniel Larocque
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8.
| |
Collapse
|
308
|
Farlow MR, Andreasen N, Riviere ME, Vostiar I, Vitaliti A, Sovago J, Caputo A, Winblad B, Graf A. Long-term treatment with active Aβ immunotherapy with CAD106 in mild Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2015; 7:23. [PMID: 25918556 PMCID: PMC4410460 DOI: 10.1186/s13195-015-0108-3] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 02/19/2015] [Indexed: 12/18/2022]
Abstract
Introduction CAD106 is designed to stimulate amyloid-β (Aβ)-specific antibody responses while avoiding T-cell autoimmune responses. The CAD106 first-in-human study demonstrated a favorable safety profile and promising antibody response. We investigated long-term safety, tolerability and antibody response after repeated CAD106 injections. Methods Two phase IIa, 52-week, multicenter, randomized, double-blind, placebo-controlled core studies (2201; 2202) and two 66-week open-label extension studies (2201E; 2202E) were conducted in patients with mild Alzheimer’s disease (AD) aged 40 to 85 years. Patients were randomized to receive 150μg CAD106 or placebo given as three subcutaneous (2201) or subcutaneous/intramuscular (2202) injections, followed by four injections (150 μg CAD106; subcutaneous, 2201E1; intramuscular, 2202E1). Our primary objective was to evaluate the safety and tolerability of repeated injections, including monitoring cerebral magnetic resonance imaging scans, adverse events (AEs) and serious AEs (SAEs). Further objectives were to assess Aβ-specific antibody response in serum and Aβ-specific T-cell response (core only). Comparable Aβ-immunoglobulin G (IgG) exposure across studies supported pooled immune response assessments. Results Fifty-eight patients were randomized (CAD106, n = 47; placebo, n = 11). Baseline demographics and characteristics were balanced. Forty-five patients entered extension studies. AEs occurred in 74.5% of CAD106-treated patients versus 63.6% of placebo-treated patients (core), and 82.2% experienced AEs during extension studies. Most AEs were mild to moderate in severity, were not study medication-related and did not require discontinuation. SAEs occurred in 19.1% of CAD106-treated patients and 36.4% of placebo-treated patients (core). One patient (CAD106-treated; 2201) reported a possibly study drug-related SAE of intracerebral hemorrhage. Four patients met criteria for amyloid-related imaging abnormalities (ARIA) corresponding to microhemorrhages: one was CAD106-treated (2201), one placebo-treated (2202) and two open-label CAD106-treated. No ARIA corresponded to vasogenic edema. Two patients discontinued extension studies because of SAEs (rectal neoplasm and rapid AD progression, respectively). Thirty CAD106-treated patients (63.8%) were serological responders. Sustained Aβ-IgG titers and prolonged time to decline were observed in extensions versus core studies. Neither Aβ1–6 nor Aβ1–42 induced specific T-cell responses; however, positive control responses were consistently detected with the CAD106 carrier. Conclusions No unexpected safety findings or Aβ-specific T-cell responses support the CAD106 favorable tolerability profile. Long-term treatment-induced Aβ-specific antibody titers and prolonged time to decline indicate antibody exposure may increase with additional injections. CAD106 may be a valuable therapeutic option in AD. Trial registration ClinicalTrials.gov identifiers: NCT00733863, registered 8 August 2008; NCT00795418, registered 10 November 2008; NCT00956410, registered 10 August 2009; NCT01023685, registered 1 December 2009. Electronic supplementary material The online version of this article (doi:10.1186/s13195-015-0108-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Martin R Farlow
- Department of Neurology, Indiana University School of Medicine, 355 West 16th Street, Suite 4700, Indianapolis, IN 46202 USA
| | - Niels Andreasen
- Karolinska Institutet, Dept NVS, Center for Alzheimer Research, Division for Neurogeriatrics, Novum, Huddinge, SE-141 57 Stockholm Sweden ; Karolinska University Hospital Huddinge, Geriatric Clinic, Clinical Trial Unit, SE-141 86 Stockholm, Sweden
| | | | | | | | | | | | - Bengt Winblad
- Karolinska Institutet, Dept NVS, Center for Alzheimer Research, Division for Neurogeriatrics, Novum, Huddinge, SE-141 57 Stockholm Sweden ; Karolinska University Hospital Huddinge, Geriatric Clinic, Clinical Trial Unit, SE-141 86 Stockholm, Sweden
| | - Ana Graf
- Novartis Pharma AG, Basel, CH-4002 Switzerland
| |
Collapse
|
309
|
Gomez-Nicola D, Boche D. Post-mortem analysis of neuroinflammatory changes in human Alzheimer's disease. Alzheimers Res Ther 2015; 7:42. [PMID: 25904988 PMCID: PMC4405851 DOI: 10.1186/s13195-015-0126-1] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Since the genome-wide association studies in Alzheimer's disease have highlighted inflammation as a driver of the disease rather than a consequence of the ongoing neurodegeneration, numerous studies have been performed to identify specific immune profiles associated with healthy, ageing, or diseased brain. However, these studies have been performed mainly in in vitro or animal models, which recapitulate only some aspects of the pathophysiology of human Alzheimer's disease. In this review, we discuss the availability of human post-mortem tissue through brain banks, the limitations associated with its use, the technical tools available, and the neuroimmune aspects to explore in order to validate in the human brain the experimental observations arising from animal models.
Collapse
Affiliation(s)
- Diego Gomez-Nicola
- />Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD UK
| | - Delphine Boche
- />Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 6YD UK
| |
Collapse
|
310
|
Murakami K. Conformation-specific antibodies to target amyloid β oligomers and their application to immunotherapy for Alzheimer's disease. Biosci Biotechnol Biochem 2015; 78:1293-305. [PMID: 25130729 DOI: 10.1080/09168451.2014.940275] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Amyloid β-protein (Aβ) oligomers, intermediates of Aβ aggregation, cause cognitive impairment and synaptotoxicity in the pathogenesis of Alzheimer's disease (AD). Immunotherapy using anti-Aβ antibody is one of the most promising approaches for AD treatment. However, most clinical trials using conventional sequence-specific antibodies have proceeded with difficulty. This is probably due to the unintended removal of the non-pathological monomer and fibrils of Aβ as well as the pathological oligomers by these antibodies that recognize Aβ sequence, which is not involved in synaptotoxicity. Several efforts have been made recently to develop conformation-specific antibodies that target the tertiary structure of Aβ oligomers. Here, we review the recent findings of Aβ oligomers and anti-Aβ antibodies including our own, and discuss their potential as therapeutic and diagnostic tools.
Collapse
Affiliation(s)
- Kazuma Murakami
- a Division of Food Science and Biotechnology , Graduate School of Agriculture, Kyoto University , Kyoto , Japan
| |
Collapse
|
311
|
Ahmed AIA, van der Marck MA, van den Elsen GAH, Olde Rikkert MGM. Cannabinoids in late-onset Alzheimer's disease. Clin Pharmacol Ther 2015; 97:597-606. [DOI: 10.1002/cpt.117] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 03/12/2015] [Indexed: 12/31/2022]
Affiliation(s)
- AIA Ahmed
- Department of Psychogeriatric Medicine; Vincent van Gogh Institute; Venray The Netherlands
- Department of Geriatric Medicine, Radboud Alzheimer Centre, and Radboud Institute for Health Sciences; Radboud University Medical Center; Nijmegen The Netherlands
- Department of Pharmacology and Toxicology; Radboud University Medical Center; Nijmegen The Netherlands
| | - MA van der Marck
- Department of Geriatric Medicine, Radboud Alzheimer Centre, and Radboud Institute for Health Sciences; Radboud University Medical Center; Nijmegen The Netherlands
| | - GAH van den Elsen
- Department of Geriatric Medicine, Radboud Alzheimer Centre, and Radboud Institute for Health Sciences; Radboud University Medical Center; Nijmegen The Netherlands
- Donders Institute for Brain Cognition and Behaviour; Radboud University Medical Center; Nijmegen The Netherlands
| | - MGM Olde Rikkert
- Department of Geriatric Medicine, Radboud Alzheimer Centre, and Radboud Institute for Health Sciences; Radboud University Medical Center; Nijmegen The Netherlands
- Donders Institute for Brain Cognition and Behaviour; Radboud University Medical Center; Nijmegen The Netherlands
| |
Collapse
|
312
|
Affiliation(s)
- Michael C Woodward
- Aged & Residential Care Services; Heidelberg Repatriation Hospital; Heidelberg Victoria
| |
Collapse
|
313
|
de la Torre Jack C. Do We Try Mending Humpty Dumpty or Prevent His Fall? An Alzheimer's Disease Dilemma. J Alzheimers Dis 2015; 46:289-96. [PMID: 25777513 DOI: 10.3233/jad-150124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In the popular nursery rhyme, Humpty Dumpty's great fall and the inability to put him together again has been used to demonstrate the second law of thermodynamics. An oversimplification of this law states that all things in the universe tend to move from order to disorder, an occurrence that can be applied allegorically to the development and clinical outcome of Alzheimer's disease (AD). An important argument relevant to the future use of resources and primary focus of AD research arises from the question, do we make it a priority to mend the shattered brain of AD patients or attempt to prevent the brain from shattering? If the former approach continues to be the priority it has become, how exactly do we mend the irreparable neuronal loss and associated cognitive failure in advanced cases of AD? Or, must we change direction and make prevention the primary goal of AD research? The latter approach would identify asymptomatic or mildly symptomatic patients with high risk of developing dementia by means of establishing multidisciplinary heart-brain clinics that would provide either close observation or a tailored therapeutic intervention. This is an important challenge that needs to be achieved if the AD incidence, societal costs and suffering, is to be significantly reduced.
Collapse
|
314
|
Rosenthal KS, Mikecz K, Steiner HL, Glant TT, Finnegan A, Carambula RE, Zimmerman DH. Rheumatoid arthritis vaccine therapies: perspectives and lessons from therapeutic ligand epitope antigen presentation system vaccines for models of rheumatoid arthritis. Expert Rev Vaccines 2015; 14:891-908. [PMID: 25787143 DOI: 10.1586/14760584.2015.1026330] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The current status of therapeutic vaccines for autoimmune diseases is reviewed with rheumatoid arthritis as the focus. Therapeutic vaccines for autoimmune diseases must regulate or subdue responses to common self-antigens. Ideally, such a vaccine would initiate an antigen-specific modulation of the T-cell immune response that drives the inflammatory disease. Appropriate animal models and types of T helper cells and signature cytokine responses that drive autoimmune disease are also discussed. Interpretation of these animal models must be done cautiously because the means of initiation, autoantigens, and even the signature cytokine and T helper cell (Th1 or Th17) responses that are involved in the disease may differ significantly from those in humans. We describe ligand epitope antigen presentation system vaccine modulation of T-cell autoimmune responses as a strategy for the design of therapeutic vaccines for rheumatoid arthritis, which may also be effective in other autoimmune conditions.
Collapse
|
315
|
Bohm C, Chen F, Sevalle J, Qamar S, Dodd R, Li Y, Schmitt-Ulms G, Fraser PE, St George-Hyslop PH. Current and future implications of basic and translational research on amyloid-β peptide production and removal pathways. Mol Cell Neurosci 2015; 66:3-11. [PMID: 25748120 PMCID: PMC4503820 DOI: 10.1016/j.mcn.2015.02.016] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 02/26/2015] [Accepted: 02/27/2015] [Indexed: 01/12/2023] Open
Abstract
Inherited variants in multiple different genes are associated with increased risk for Alzheimer's disease (AD). In many of these genes, the inherited variants alter some aspect of the production or clearance of the neurotoxic amyloid β-peptide (Aβ). Thus missense, splice site or duplication mutants in the presenilin 1 (PS1), presenilin 2 (PS2) or the amyloid precursor protein (APP) genes, which alter the levels or shift the balance of Aβ produced, are associated with rare, highly penetrant autosomal dominant forms of Familial Alzheimer's Disease (FAD). Similarly, the more prevalent late-onset forms of AD are associated with both coding and non-coding variants in genes such as SORL1, PICALM and ABCA7 that affect the production and clearance of Aβ. This review summarises some of the recent molecular and structural work on the role of these genes and the proteins coded by them in the biology of Aβ. We also briefly outline how the emerging knowledge about the pathways involved in Aβ generation and clearance can be potentially targeted therapeutically. This article is part of Special Issue entitled "Neuronal Protein".
Collapse
Affiliation(s)
- C Bohm
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - F Chen
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - J Sevalle
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - S Qamar
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrookes Hospital, Hills Road, Cambridge CB2 0XY, UK
| | - R Dodd
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrookes Hospital, Hills Road, Cambridge CB2 0XY, UK
| | - Y Li
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrookes Hospital, Hills Road, Cambridge CB2 0XY, UK
| | - G Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - P E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - P H St George-Hyslop
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada; Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrookes Hospital, Hills Road, Cambridge CB2 0XY, UK.
| |
Collapse
|
316
|
Verdier JM, Acquatella I, Lautier C, Devau G, Trouche S, Lasbleiz C, Mestre-Francés N. Lessons from the analysis of nonhuman primates for understanding human aging and neurodegenerative diseases. Front Neurosci 2015; 9:64. [PMID: 25788873 PMCID: PMC4349082 DOI: 10.3389/fnins.2015.00064] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 02/13/2015] [Indexed: 12/13/2022] Open
Abstract
Animal models are necessary tools for solving the most serious challenges facing medical research. In aging and neurodegenerative disease studies, rodents occupy a place of choice. However, the most challenging questions about longevity, the complexity and functioning of brain networks or social intelligence can almost only be investigated in nonhuman primates. Beside the fact that their brain structure is much closer to that of humans, they develop highly complex cognitive strategies and they are visually-oriented like humans. For these reasons, they deserve consideration, although their management and care are more complicated and the related costs much higher. Despite these caveats, considerable scientific advances have been possible using nonhuman primates. This review concisely summarizes their role in the study of aging and of the mechanisms involved in neurodegenerative disorders associated mainly with cognitive dysfunctions (Alzheimer's and prion diseases) or motor deficits (Parkinson's and related diseases).
Collapse
Affiliation(s)
- Jean-Michel Verdier
- Université de Montpellier Montpellier, France ; Institut National de la Santé et de la Recherche Médicale, U1198 Montpellier, France ; Ecole Pratique des Hautes Etudes Paris, France
| | - Isabelle Acquatella
- Université de Montpellier Montpellier, France ; Institut National de la Santé et de la Recherche Médicale, U1198 Montpellier, France ; Ecole Pratique des Hautes Etudes Paris, France
| | - Corinne Lautier
- Université de Montpellier Montpellier, France ; Institut National de la Santé et de la Recherche Médicale, U1198 Montpellier, France ; Ecole Pratique des Hautes Etudes Paris, France
| | - Gina Devau
- Université de Montpellier Montpellier, France ; Institut National de la Santé et de la Recherche Médicale, U1198 Montpellier, France ; Ecole Pratique des Hautes Etudes Paris, France
| | - Stéphanie Trouche
- Université de Montpellier Montpellier, France ; Institut National de la Santé et de la Recherche Médicale, U1198 Montpellier, France ; Ecole Pratique des Hautes Etudes Paris, France
| | - Christelle Lasbleiz
- Université de Montpellier Montpellier, France ; Institut National de la Santé et de la Recherche Médicale, U1198 Montpellier, France ; Ecole Pratique des Hautes Etudes Paris, France
| | - Nadine Mestre-Francés
- Université de Montpellier Montpellier, France ; Institut National de la Santé et de la Recherche Médicale, U1198 Montpellier, France ; Ecole Pratique des Hautes Etudes Paris, France
| |
Collapse
|
317
|
A comparative evaluation of a novel vaccine in APP/PS1 mouse models of Alzheimer's disease. BIOMED RESEARCH INTERNATIONAL 2015; 2015:807146. [PMID: 25759822 PMCID: PMC4339718 DOI: 10.1155/2015/807146] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 09/14/2014] [Indexed: 12/13/2022]
Abstract
Immunization against amyloid-beta-peptide (Aβ) has been widely investigated as a potential immunotherapeutic approach for Alzheimer's disease (AD). With the aim of developing an active immunogenic vaccine without need of coadjuvant modification for human trials and therefore avoiding such side effects, we designed the Aβ1–42 vaccine (EB101), delivered in a liposomal matrix, that based on our previous studies significantly prevents and reverses the AD neuropathology, clearing Aβ plaques while markedly reducing neuronal degeneration, behavioral deficits, and minimizing neuroinflammation in APP/PS1 transgenic mice. Here, the efficacy of our immunogenic vaccine EB101 was compared with the original immunization vaccine cocktail Aβ42 + CFA/IFA (Freund's adjuvant), in order to characterize the effect of sphingosine-1-phosphate (S1P) in the immunotherapeutic response. Quantitative analysis of amyloid burden showed a notable decrease in the neuroinflammation reaction against Aβ plaques when S1P was compared with other treatments, suggesting that S1P plays a key role as a neuroprotective agent. Moreover, EB101 immunized mice presented a protective immunogenic reaction resulting in the increase of Aβ-specific antibody response and decrease of reactive glia in the affected brain areas, leading to a Th2 immunological reaction.
Collapse
|
318
|
Nakagami H, Morishita R. Development of DNA vaccines as an anti-hypertensive therapy or for anti-angiogenesis. Expert Opin Biol Ther 2015; 15:431-6. [PMID: 25673364 DOI: 10.1517/14712598.2015.1014791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Vaccines are used as preventive medicine for infectious diseases worldwide; however, several recent studies have indicated the potential of therapeutic vaccines for managing Alzheimer's disease or hypertension. AREAS COVERED The concept and history of therapeutic vaccines for hypertension are introduced. The improvement of drug compliance associated with vaccines in hypertensive patients may assist in better control of blood pressure, leading to reduced complications. Recently, groups have attempted to develop a therapeutic vaccine against hypertension. The vaccine-induced anti-angiotensin II antibodies can efficiently ameliorate high blood pressure. DNA vaccines have also been designed via a similar strategy using a plasmid vector encoding a hepatitis B core (HBc)-angiotensin II fusion protein. The immunogenic molecule (i.e., HBc) is used to provide an antigen that supports the activation of T cells in this DNA vaccine system. The platform technology can also be applied to generate a VEGF vaccine for cancer. EXPERT OPINION To date, several clinical studies of DNA vaccines have been conducted, but their effectiveness has not been determined. We hope that the novel concept of DNA vaccines will contribute to promoting health and medicine in the future.
Collapse
Affiliation(s)
- Hironori Nakagami
- Osaka University, Osaka University United Graduate School of Child Development, Kanazawa University and Hamamatsu University School of Medicine, Division of Vascular Medicine and Epigenetics , 2-1 Yamada-oka, Suita, Osaka 565-0871 , Japan +81 6 6879 4142 ; +81 6 6879 4142 ;
| | | |
Collapse
|
319
|
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
|
320
|
Kou J, Yang J, Lim JE, Pattanayak A, Song M, Planque S, Paul S, Fukuchi KI. Catalytic immunoglobulin gene delivery in a mouse model of Alzheimer's disease: prophylactic and therapeutic applications. Mol Neurobiol 2015; 51:43-56. [PMID: 24733587 PMCID: PMC4198531 DOI: 10.1007/s12035-014-8691-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 03/24/2014] [Indexed: 12/12/2022]
Abstract
Accumulation of amyloid beta-peptide (Aβ) in the brain is hypothesized to be a causal event leading to dementia in Alzheimer's disease (AD). Aβ vaccination removes Aβ deposits from the brain. Aβ immunotherapy, however, may cause T cell- and/or Fc-receptor-mediated brain inflammation and relocate parenchymal Aβ deposits to blood vessels leading to cerebral hemorrhages. Because catalytic antibodies do not form stable immune complexes and Aβ fragments produced by catalytic antibodies are less likely to form aggregates, Aβ-specific catalytic antibodies may have safer therapeutic profiles than reversibly-binding anti-Aβ antibodies. Additionally, catalytic antibodies may remove Aβ more efficiently than binding antibodies because a single catalytic antibody can hydrolyze thousands of Aβ molecules. We previously isolated Aβ-specific catalytic antibody, IgVL5D3, with strong Aβ-hydrolyzing activity. Here, we evaluated the prophylactic and therapeutic efficacy of brain-targeted IgVL5D3 gene delivery via recombinant adeno-associated virus serotype 9 (rAAV9) in an AD mouse model. One single injection of rAAV9-IgVL5D3 into the right ventricle of AD model mice yielded widespread, high expression of IgVL5D3 in the unilateral hemisphere. IgVL5D3 expression was readily detectable in the contralateral hemisphere but to a much lesser extent. IgVL5D3 expression was also confirmed in the cerebrospinal fluid. Prophylactic and therapeutic injection of rAAV9-IgVL5D3 reduced Aβ load in the ipsilateral hippocampus of AD model mice. No evidence of hemorrhages, increased vascular amyloid deposits, increased proinflammatory cytokines, or infiltrating T-cells in the brains was found in the experimental animals. AAV9-mediated anti-Aβ catalytic antibody brain delivery can be prophylactic and therapeutic options for AD.
Collapse
Affiliation(s)
- Jinghong Kou
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois 61656, USA
| | - Junling Yang
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois 61656, USA
| | - Jeong-Eun Lim
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois 61656, USA
| | - Abhinandan Pattanayak
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois 61656, USA
| | - Min Song
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois 61656, USA
| | - Stephanie Planque
- Chemical Immunology Research Center, Department of Pathology and Laboratory Medicine, University of Texas Houston Medical School, Houston, Texas 77030, USA
| | - Sudhir Paul
- Chemical Immunology Research Center, Department of Pathology and Laboratory Medicine, University of Texas Houston Medical School, Houston, Texas 77030, USA
| | - Ken-ichiro Fukuchi
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois 61656, USA
| |
Collapse
|
321
|
Nisbet RM, Polanco JC, Ittner LM, Götz J. Tau aggregation and its interplay with amyloid-β. Acta Neuropathol 2015; 129:207-20. [PMID: 25492702 PMCID: PMC4305093 DOI: 10.1007/s00401-014-1371-2] [Citation(s) in RCA: 233] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Revised: 11/11/2014] [Accepted: 11/26/2014] [Indexed: 01/09/2023]
Abstract
Neurofibrillary tangles and amyloid plaques constitute the hallmark brain lesions of Alzheimer's disease (AD) patients. Tangles are composed of fibrillar aggregates of the microtubule-associated protein tau, and plaques comprise fibrillar forms of a proteolytic cleavage product, amyloid-β (Aβ). Although plaques and tangles are the end-stage lesions in AD, small oligomers of Aβ and tau are now receiving increased attention as they are shown to correlate best with neurotoxicity. One key question of debate, however, is which of these pathologies appears first and hence is upstream in the pathocascade. Studies suggest that there is an intense crosstalk between the two molecules and, based on work in animal models, there is increasing evidence that Aβ, at least in part, exerts its toxicity via tau, with the Src kinase Fyn playing a crucial role in this process. In other experimental paradigms, Aβ and tau have been found to exert both separate and synergistic modes of toxicity. The challenge, however, is to integrate these different scenarios into a coherent picture. Furthermore, the ability of therapeutic interventions targeting just one of these molecules, to successfully neutralize the toxicity of the other, needs to be ascertained to improve current therapeutic strategies, such as immunotherapy, for the treatment of AD. Although this article is not intended to provide a comprehensive review of the currently pursued therapeutic strategies, we will discuss what has been achieved by immunotherapy and, in particular, how the inherent limitations of this approach can possibly be overcome by novel strategies that involve single-chain antibodies.
Collapse
Affiliation(s)
- Rebecca M. Nisbet
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Juan-Carlos Polanco
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Lars M. Ittner
- Dementia Research Unit, Wallace Wurth Building, The University of New South Wales, Sydney, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| |
Collapse
|
322
|
Yamada M. Cerebral amyloid angiopathy: emerging concepts. J Stroke 2015; 17:17-30. [PMID: 25692104 PMCID: PMC4325636 DOI: 10.5853/jos.2015.17.1.17] [Citation(s) in RCA: 249] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 12/15/2014] [Accepted: 12/24/2014] [Indexed: 12/15/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA) involves cerebrovascular amyloid deposition and is classified into several types according to the amyloid protein involved. Of these, sporadic amyloid β-protein (Aβ)-type CAA is most commonly found in older individuals and in patients with Alzheimer's disease (AD). Cerebrovascular Aβ deposits accompany functional and pathological changes in cerebral blood vessels (CAA-associated vasculopathies). CAA-associated vasculopathies lead to development of hemorrhagic lesions [lobar intracerebral macrohemorrhage, cortical microhemorrhage, and cortical superficial siderosis (cSS)/focal convexity subarachnoid hemorrhage (SAH)], ischemic lesions (cortical infarction and ischemic changes of the white matter), and encephalopathies that include subacute leukoencephalopathy caused by CAA-associated inflammation/angiitis. Thus, CAA is related to dementia, stroke, and encephalopathies. Recent advances in diagnostic procedures, particularly neuroimaging, have enabled us to establish a clinical diagnosis of CAA without brain biopsies. Sensitive magnetic resonance imaging (MRI) methods, such as gradient-echo T2* imaging and susceptibility-weighted imaging, are useful for detecting cortical microhemorrhages and cSS. Amyloid imaging with amyloid-binding positron emission tomography (PET) ligands, such as Pittsburgh Compound B, can detect CAA, although they cannot discriminate vascular from parenchymal amyloid deposits. In addition, cerebrospinal fluid markers may be useful, including levels of Aβ40 for CAA and anti-Aβ antibody for CAA-related inflammation. Moreover, cSS is closely associated with transient focal neurological episodes (TFNE). CAA-related inflammation/angiitis shares pathophysiology with amyloid-related imaging abnormalities (ARIA) induced by Aβ immunotherapies in AD patients. This article reviews CAA and CAA-related disorders with respect to their epidemiology, pathology, pathophysiology, clinical features, biomarkers, diagnosis, treatment, risk factors, and future perspectives.
Collapse
Affiliation(s)
- Masahito Yamada
- Department of Neurology and Neurobiology of Aging, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| |
Collapse
|
323
|
Takagane K, Nojima J, Mitsuhashi H, Suo S, Yanagihara D, Takaiwa F, Urano Y, Noguchi N, Ishiura S. Aβ induces oxidative stress in senescence-accelerated (SAMP8) mice. Biosci Biotechnol Biochem 2015; 79:912-8. [PMID: 25612552 DOI: 10.1080/09168451.2014.1002449] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
According to the amyloid hypothesis, amyloid β accumulates in brains with Alzheimer's disease (AD) and triggers cell death and memory deficit. Previously, we developed a rice Aβ vaccine expressing Aβ, which reduced brain Aβ levels in the Tg2576 mouse model of familial AD. We used senescence-accelerated SAMP8 mice as a model of sporadic AD and investigated the relationship between Aβ and oxidative stress. Insoluble Aβ and 4-hydroxynonenal (4-HNE) levels tended to be reduced in SAMP8 mice-fed the rice Aβ vaccine. We attempted to clarify the relationship between oxidative stress and Aβ in vitro. Addition of Aβ peptide to the culture medium resulted in an increase in 4-HNE levels in SH-SY5Y cells. Tg2576 mice, which express large amounts of Aβ in their brain, also exhibited increased 4-HNE levels; this increase was inhibited by the Aβ vaccine. These results indicate that Aβ induces oxidative stress in cultured cells and in the mouse brain.
Collapse
Affiliation(s)
- Kurara Takagane
- a Department of Biological Sciences, Graduate School of Science , The University of Tokyo , Tokyo , Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
324
|
Mandler M, Santic R, Gruber P, Cinar Y, Pichler D, Funke SA, Willbold D, Schneeberger A, Schmidt W, Mattner F. Tailoring the antibody response to aggregated Aß using novel Alzheimer-vaccines. PLoS One 2015; 10:e0115237. [PMID: 25611858 PMCID: PMC4303436 DOI: 10.1371/journal.pone.0115237] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 11/20/2014] [Indexed: 12/26/2022] Open
Abstract
Recent evidence suggests Alzheimer-Disease (AD) to be driven by aggregated Aß. Capitalizing on the mechanism of molecular mimicry and applying several selection layers, we screened peptide libraries for moieties inducing antibodies selectively reacting with Aß-aggregates. The technology identified a pool of peptide candidates; two, AFFITOPES AD01 and AD02, were assessed as vaccination antigens and compared to Aβ1-6, the targeted epitope. When conjugated to Keyhole Limpet Hemocyanin (KLH) and adjuvanted with aluminum, all three peptides induced Aß-targeting antibodies (Abs). In contrast to Aß1-6, AD01- or AD02-induced Abs were characterized by selectivity for aggregated forms of Aß and absence of reactivity with related molecules such as Amyloid Precursor Protein (APP)/ secreted APP-alpha (sAPPa). Administration of AFFITOPE-vaccines to APP-transgenic mice was found to reduce their cerebral amyloid burden, the associated neuropathological alterations and to improve their cognitive functions. Thus, the AFFITOME-technology delivers vaccines capable of inducing a distinct Ab response. Their features may be beneficial to AD-patients, a hypothesis currently tested within a phase-II-study.
Collapse
Affiliation(s)
- Markus Mandler
- AFFiRiS AG, Karl-Farkas-Gasse 22, A-1030, Vienna, Austria
| | - Radmila Santic
- AFFiRiS AG, Karl-Farkas-Gasse 22, A-1030, Vienna, Austria
| | - Petra Gruber
- AFFiRiS AG, Karl-Farkas-Gasse 22, A-1030, Vienna, Austria
| | - Yeliz Cinar
- Institute for Structural Biochemistry (Institute of Complex Systems 6), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Dagmar Pichler
- AFFiRiS AG, Karl-Farkas-Gasse 22, A-1030, Vienna, Austria
| | - Susanne Aileen Funke
- Institute for Structural Biochemistry (Institute of Complex Systems 6), Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Dieter Willbold
- Institute for Structural Biochemistry (Institute of Complex Systems 6), Forschungszentrum Jülich, 52425, Jülich, Germany
| | | | - Walter Schmidt
- AFFiRiS AG, Karl-Farkas-Gasse 22, A-1030, Vienna, Austria
| | - Frank Mattner
- AFFiRiS AG, Karl-Farkas-Gasse 22, A-1030, Vienna, Austria
| |
Collapse
|
325
|
|
326
|
Co-immunization with DNA and protein mixture: a safe and efficacious immunotherapeutic strategy for Alzheimer's disease in PDAPP mice. Sci Rep 2015; 5:7771. [PMID: 25586780 PMCID: PMC4293606 DOI: 10.1038/srep07771] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 12/03/2014] [Indexed: 11/15/2022] Open
Abstract
Active immunotherapy targeting β-amyloid (Aβ) is the most promising strategy to prevent or treat Alzheimer's disease (AD). Based on pre-clinical studies and clinical trials, a safe and effective AD vaccine requires a delicate balance between providing therapeutically adequate anti-Aβ antibodies and eliminating or suppressing unwanted adverse T cell-mediated inflammatory reactions. We describe here the immunological characterization and protective efficacy of co-immunization with a 6Aβ15-T DNA and protein mixture without adjuvant as an AD immunotherapeutic strategy. Impressively, this co-immunization induced robust Th2-polarized Aβ-specific antibodies while simultaneously suppressed unwanted inflammatory T cell reactions and avoiding Aβ42-specific T cell-mediated autoimmune responses in immunized mice. Co-immunization with the DNA + protein vaccine could overcome Aβ42-associated hypo-responsiveness and elicit long-term Aβ-specific antibody responses, which helped to maintain antibody-mediated clearance of amyloid and accordingly alleviated AD symptoms in co-immunized PDAPP mice. Our DNA and protein combined vaccine, which could induce an anti-inflammatory Th2 immune response with high level Aβ-specific antibodies and low level IFN-γ production, also demonstrated the capacity to inhibit amyloid accumulation and prevent cognitive dysfunction. Hence, co-immunization with antigen-matched DNA and protein may represent a novel and efficacious strategy for AD immunotherapy to eliminate T cell inflammatory reactions while retaining high level antibody responses.
Collapse
|
327
|
Weathers SP, Gilbert MR. Current challenges in designing GBM trials for immunotherapy. J Neurooncol 2015; 123:331-7. [DOI: 10.1007/s11060-015-1716-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 12/31/2014] [Indexed: 12/21/2022]
|
328
|
Kohyama K, Matsumoto Y. Alzheimer's disease and immunotherapy: what is wrong with clinical trials? Immunotargets Ther 2015; 4:27-34. [PMID: 27471709 PMCID: PMC4918245 DOI: 10.2147/itt.s49923] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by progressive neurodegeneration and is the most common cause of dementia. Immunotherapy has recently been regarded as a potential treatment for AD. This stems from the fact that the clinical and pathological findings from the active AD vaccine trial suggests that such vaccine therapy may be effective for AD. However, this trial was halted because of the occurrence of meningoencephalitis in some patients. Avoiding excessive immune reaction is necessary for the success of vaccine therapy. For this purpose, adjuvant-free vaccine therapies (eg, passive immunization or DNA vaccines) are currently under investigation. However, the results of clinical trials employing both active and passive anti-amyloid-beta immunotherapy have been unsatisfactory. In this article, we will analyze the reasons for the limited efficacy of currently available immunotherapies and discuss the effectiveness of new vaccine therapies. Finally, we will speculate on the possibility of its clinical application.
Collapse
Affiliation(s)
- Kuniko Kohyama
- Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yoh Matsumoto
- Department of Sensory and Motor Systems, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan; Immunotherapy Development Inc., Saitama, Japan; Geriatric Health Services Facility "Asahigaoka", Saitama, Japan
| |
Collapse
|
329
|
Paquet C, Amin J, Mouton-Liger F, Nasser M, Love S, Gray F, Pickering RM, Nicoll JAR, Holmes C, Hugon J, Boche D. Effect of active Aβ immunotherapy on neurons in human Alzheimer's disease. J Pathol 2015; 235:721-30. [PMID: 25430817 DOI: 10.1002/path.4491] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 10/29/2014] [Accepted: 11/26/2014] [Indexed: 12/26/2022]
Abstract
Amyloid β peptide (Aβ) immunization of Alzheimer's disease (AD) patients has been reported to induce amyloid plaque removal, but with little impact on cognitive decline. We have explored the consequences of Aβ immunotherapy on neurons in post mortem brain tissue. Eleven immunized (AN1792, Elan Pharmaceuticals) AD patients were compared to 28 non-immunized AD cases. Immunohistochemistry on sections of neocortex was performed for neuron-specific nuclear antigen (NeuN), neurofilament protein (NFP) and phosphorylated-(p)PKR (pro-apoptotic kinase detected in degenerating neurons). Quantification was performed for pPKR and status spongiosis (neuropil degeneration), NeuN-positive neurons/field, curvature of the neuronal processes and interneuronal distance. Data were corrected for age, gender, duration of dementia and APOE genotype and also assessed in relation to Aβ42 and tau pathology and key features of AD. In non-immunized patients, the degree of neuritic curvature correlated with spongiosis and pPKR, and overall the neurodegenerative markers correlated better with tau pathology than Aβ42 load. Following immunization, spongiosis increased, interneuronal distance increased, while the number of NeuN-positive neurons decreased, consistent with enhanced neuronal loss. However, neuritic curvature was reduced and pPKR was associated with Aβ removal in immunized patients. In AD, associations of spongiosis status, curvature ratio and pPKR load with microglial markers Iba1, CD68 and CD32 suggest a role for microglia in neurodegeneration. After immunization, correlations were detected between the number of NeuN-positive neurons and pPKR with Iba1, CD68 and CD64, suggesting that microglia are involved in the neuronal loss. Our findings suggest that in established AD this form of active Aβ immunization may predominantly accelerate loss of damaged degenerating neurons. This interpretation is consistent with in vivo imaging indicating an increased rate of cerebral atrophy in immunized AD patients.
Collapse
Affiliation(s)
- Claire Paquet
- Alzheimer Clinical Centre, Lariboisiere FW Saint-Louis Hospital, AP-HP University of Paris Diderot, France; Department of Histology and Biology of Ageing, Lariboisiere FW Saint-Louis Hospital, AP-HP University of Paris Diderot, France; INSERM U942, Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
330
|
Medina M, Avila J. Further understanding of tau phosphorylation: implications for therapy. Expert Rev Neurother 2015; 15:115-22. [PMID: 25555397 DOI: 10.1586/14737175.2015.1000864] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Tau is a brain microtubule-associated protein that regulates microtubule structure and function. Prominent tau neurofibrillary pathology is a common feature in a number of neurodegenerative disorders collectively referred to as tauopathies, the most common of which is Alzheimer's disease. Beyond its classical role as a microtubule-associated protein, recent advances in our understanding of tau cellular functions have unveiled novel important tau cellular functions that may also play a pivotal role in pathogenesis and render novel targets for therapeutic intervention. Regulation of tau behavior and function under physiological and pathological conditions is mainly achieved through post-translational modifications, especially phosphorylation, which has significant implications for the development of novel therapeutic approaches in a number of neurodegenerative disorders.
Collapse
Affiliation(s)
- Miguel Medina
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Valderrebollo 5, 28041-Madrid, Spain
| | | |
Collapse
|
331
|
Rafii MS. Active Immunotherapy for Alzheimer's Disease: The Road Ahead. JPAD-JOURNAL OF PREVENTION OF ALZHEIMERS DISEASE 2015; 2:78-79. [PMID: 28331846 DOI: 10.14283/jpad.2015.59] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- M S Rafii
- Department of Neurosciences, University of California, San Diego
| |
Collapse
|
332
|
Robinson M, Yasie Lee B, Leonenko Z. Drugs and drug delivery systems targeting amyloid-β in Alzheimer's disease. AIMS MOLECULAR SCIENCE 2015. [DOI: 10.3934/molsci.2015.3.332] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
333
|
Kang JH, Ryoo NY, Shin DW, Trojanowski JQ, Shaw LM. Role of cerebrospinal fluid biomarkers in clinical trials for Alzheimer's disease modifying therapies. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2014; 18:447-56. [PMID: 25598657 PMCID: PMC4296032 DOI: 10.4196/kjpp.2014.18.6.447] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 09/02/2014] [Accepted: 10/07/2014] [Indexed: 12/27/2022]
Abstract
Until now, a disease-modifying therapy (DMT) that has an ability to slow or arrest Alzheimer's disease (AD) progression has not been developed, and all clinical trials involving AD patients enrolled by clinical assessment alone also have not been successful. Given the growing consensus that the DMT is likely to require treatment initiation well before full-blown dementia emerges, the early detection of AD will provide opportunities to successfully identify new drugs that slow the course of AD pathology. Recent advances in early detection of AD and prediction of progression of the disease using various biomarkers, including cerebrospinal fluid (CSF) Aβ1-42, total tau and p-tau181 levels, and imagining biomarkers, are now being actively integrated into the designs of AD clinical trials. In terms of therapeutic mechanisms, monitoring these markers may be helpful for go/no-go decision making as well as surrogate markers for disease severity or progression. Furthermore, CSF biomarkers can be used as a tool to enrich patients for clinical trials with prospect of increasing statistical power and reducing costs in drug development. However, the standardization of technical aspects of analysis of these biomarkers is an essential prerequisite to the clinical uses. To accomplish this, global efforts are underway to standardize CSF biomarker measurements and a quality control program supported by the Alzheimer's Association. The current review summarizes therapeutic targets of developing drugs in AD pathophysiology, and provides the most recent advances in the
Collapse
Affiliation(s)
- Ju-Hee Kang
- Department of Pharmacology and Clinical Pharmacology, Inha University School of Medicine, Incheon 400-712, Korea. ; Hypoxia-related Disease Research Center, Inha University School of Medicine, Incheon 400-712, Korea. ; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Na-Young Ryoo
- Hypoxia-related Disease Research Center, Inha University School of Medicine, Incheon 400-712, Korea. ; Department of Anatomy, Inha University School of Medicine, Incheon 400-712, Korea
| | - Dong Wun Shin
- Department of Emergency Medicine, Inje University Ilsan Paik Hospital, Ilsan 411-706, Korea
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA. ; Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA. ; Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
334
|
Neus Bosch M, Pugliese M, Andrade C, Gimeno-Bay�n J, Mahy N, Rodriguez MJ. Amyloid-� Immunotherapy Reduces Amyloid Plaques and Astroglial Reaction in Aged Domestic Dogs. NEURODEGENER DIS 2014; 15:24-37. [DOI: 10.1159/000368672] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 09/25/2014] [Indexed: 11/19/2022] Open
|
335
|
Cash DM, Rohrer JD, Ryan NS, Ourselin S, Fox NC. Imaging endpoints for clinical trials in Alzheimer's disease. Alzheimers Res Ther 2014; 6:87. [PMID: 25621018 PMCID: PMC4304258 DOI: 10.1186/s13195-014-0087-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
As the need to develop a successful disease-modifying treatment for Alzheimer's disease (AD) becomes more urgent, imaging is increasingly used in therapeutic trials. We provide an overview of how the different imaging modalities are used in AD studies and the current regulatory guidelines for their use in clinical trials as endpoints. We review the current literature for results of imaging endpoints of efficacy and safety in published clinical trials. We start with trials in mild to moderate AD, where imaging (largely magnetic resonance imaging (MRI)) has long played a role in inclusion and exclusion criteria; more recently, MRI has been used to identify adverse events and to measure rates of brain atrophy. The advent of amyloid imaging using positron emission tomography has led to trials incorporating amyloid measurements as endpoints and incidentally to the recognition of the high proportion of amyloid-negative individuals that may be recruited into these trials. Ongoing and planned trials now commonly include multimodality imaging: amyloid positron emission tomography, MRI and other modalities. At the same time, the failure of recent large profile trials in mild to moderate AD together with the realisation that there is a long prodromal period to AD has driven a push to move studies to earlier in the disease. Imaging has particularly important roles, alongside other biomarkers, in assessing efficacy because conventional clinical outcomes may have limited ability to detect treatment effects in these early stages.
Collapse
Affiliation(s)
- David M Cash
- />Dementia Research Centre, Box 16, The National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG UK
- />Translational Imaging Group, Centre for Medical Image Computing, University College of London, 3rd Floor, Wolfson House, 4 Stephenson Way, London, NW1 2HE UK
| | - Jonathan D Rohrer
- />Dementia Research Centre, Box 16, The National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG UK
| | - Natalie S Ryan
- />Dementia Research Centre, Box 16, The National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG UK
| | - Sebastien Ourselin
- />Dementia Research Centre, Box 16, The National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG UK
- />Translational Imaging Group, Centre for Medical Image Computing, University College of London, 3rd Floor, Wolfson House, 4 Stephenson Way, London, NW1 2HE UK
| | - Nick C Fox
- />Dementia Research Centre, Box 16, The National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG UK
| |
Collapse
|
336
|
Davtyan H, Ghochikyan A, Hovakimyan A, Davtyan A, Cadagan R, Marleau AM, Albrecht RA, García-Sastre A, Agadjanyan MG. A dual vaccine against influenza & Alzheimer's disease failed to enhance anti-β-amyloid antibody responses in mice with pre-existing virus specific memory. J Neuroimmunol 2014; 277:77-84. [PMID: 25455094 PMCID: PMC4314405 DOI: 10.1016/j.jneuroim.2014.10.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 10/03/2014] [Accepted: 10/07/2014] [Indexed: 01/13/2023]
Abstract
Novel dual vaccine, WSN-Aβ(1-10), based on the recombinant influenza virus, expressing immunodominant B-cell epitope of β-amyloid, simultaneously induced therapeutically potent anti-Aβ and anti-influenza antibodies. In this study we showed that boosting of WSN-WT primed mice with WSN-Aβ(1-10) enhances anti-viral, but fails to induce anti-Aβ antibody responses. This inhibition is associated with expression of Aβ(1-10) within the context of an inactivated influenza virus vaccine. These results demonstrate that the use of an inactivated influenza virus as a carrier for AD vaccine may not be applicable due to the possible inhibition of anti-Aβ antibody response in individuals previously vaccinated or infected with influenza.
Collapse
Affiliation(s)
- Hayk Davtyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA
| | - Anahit Ghochikyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA
| | - Armine Hovakimyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA
| | - Arpine Davtyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA
| | - Richard Cadagan
- Department of Microbiology, Mount Sinai School of Medicine, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Annette M Marleau
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA
| | - Randy A Albrecht
- Department of Microbiology, Mount Sinai School of Medicine, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Mount Sinai School of Medicine, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Mount Sinai School of Medicine, New York, NY 10029, USA; Department of Medicine, Division of Infection Diseases, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Michael G Agadjanyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; University of California, Irvine, Institute for Memory Impairments and Neurological Disorders, Irvine, CA 92697, USA.
| |
Collapse
|
337
|
Lambracht-Washington D, Rosenberg RN. A noninflammatory immune response in aged DNA Aβ42-immunized mice supports its safety for possible use as immunotherapy in AD patients. Neurobiol Aging 2014; 36:1274-81. [PMID: 25725942 DOI: 10.1016/j.neurobiolaging.2014.12.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/02/2014] [Accepted: 12/06/2014] [Indexed: 12/17/2022]
Abstract
Aging in the immune system results in tendency to proinflammatory responses. Intradermal DNA immunization showed Th2 polarized noninflammatory immune responses. We tested here 18-month-old mice which were immunized with Aβ42 peptide, DNA Aβ42 trimer, or 2 different prime boost protocols identical to previous experiments. High Aβ42 antibody levels were found in aged mice which had received peptide immunizations (900 μg/mL plasma), and in mice which had received peptide prime and DNA boost immunizations (500 μg/mL), compared with antibodies in DNA Aβ42 immunized mice with 50 μg/mL. Although we found T-cell proliferation and inflammatory cytokines in mice which had received peptide or prime boost immunization, these were not found in DNA-immunized mice. The results are concordant with proinflammatory responses because of immunosenescence and contraindicate the use of Aβ42 peptide immunizations or prime boost immunization protocols for the use in elderly Alzheimer's disease patients. DNA Aβ42 immunization only on the other hand does lead to effective levels of antibodies without inflammatory cytokine or T-cell responses in the aged animal model tested.
Collapse
Affiliation(s)
- Doris Lambracht-Washington
- Department of Neurology and Neurotherapeutics, Alzheimer's Disease Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Roger N Rosenberg
- Department of Neurology and Neurotherapeutics, Alzheimer's Disease Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
338
|
Kilic E, Reitmeir R, Kilic Ü, Caglayan AB, Beker MC, Kelestemur T, Ethemoglu MS, Ozturk G, Hermann DM. HMG-CoA Reductase Inhibition Promotes Neurological Recovery, Peri-Lesional Tissue Remodeling, and Contralesional Pyramidal Tract Plasticity after Focal Cerebral Ischemia. Front Cell Neurosci 2014; 8:422. [PMID: 25565957 PMCID: PMC4263103 DOI: 10.3389/fncel.2014.00422] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 11/23/2014] [Indexed: 01/17/2023] Open
Abstract
3-Hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors are widely used for secondary stroke prevention. Besides their lipid-lowering activity, pleiotropic effects on neuronal survival, angiogenesis, and neurogenesis have been described. In view of these observations, we were interested whether HMG-CoA reductase inhibition in the post-acute stroke phase promotes neurological recovery, peri-lesional, and contralesional neuronal plasticity. We examined effects of the HMG-CoA reductase inhibitor rosuvastatin (0.2 or 2.0 mg/kg/day i.c.v.), administered starting 3 days after 30 min of middle cerebral artery occlusion for 30 days. Here, we show that rosuvastatin treatment significantly increased the grip strength and motor coordination of animals, promoted exploration behavior, and reduced anxiety. It was associated with structural remodeling of peri-lesional brain tissue, reflected by increased neuronal survival, enhanced capillary density, and reduced striatal and corpus callosum atrophy. Increased sprouting of contralesional pyramidal tract fibers crossing the midline in order to innervate the ipsilesional red nucleus was noticed in rosuvastatin compared with vehicle-treated mice, as shown by anterograde tract tracing experiments. Western blot analysis revealed that the abundance of HMG-CoA reductase was increased in the contralesional hemisphere at 14 and 28 days post-ischemia. Our data support the idea that HMG-CoA reductase inhibition promotes brain remodeling and plasticity far beyond the acute stroke phase, resulting in neurological recovery.
Collapse
Affiliation(s)
- Ertugrul Kilic
- Department of Physiology, Istanbul Medipol University , Istanbul , Turkey
| | - Raluca Reitmeir
- Department of Neurology, University Hospital , Essen , Germany
| | - Ülkan Kilic
- Department of Medical Biology, Istanbul Medipol University , Istanbul , Turkey
| | | | | | - Taha Kelestemur
- Department of Physiology, Istanbul Medipol University , Istanbul , Turkey
| | | | - Gurkan Ozturk
- Department of Physiology, Istanbul Medipol University , Istanbul , Turkey
| | - Dirk M Hermann
- Department of Neurology, University Hospital , Essen , Germany
| |
Collapse
|
339
|
Lambracht-Washington D, Rosenberg RN. Co-stimulation with TNF receptor superfamily 4/25 antibodies enhances in-vivo expansion of CD4+CD25+Foxp3+ T cells (Tregs) in a mouse study for active DNA Aβ42 immunotherapy. J Neuroimmunol 2014; 278:90-9. [PMID: 25595257 DOI: 10.1016/j.jneuroim.2014.12.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/05/2014] [Accepted: 12/08/2014] [Indexed: 01/09/2023]
Abstract
The study was designed to test DNA Aβ42 immunization in mice as alternative approach for possible active immunotherapy in Alzheimer patients. As results, we found polarized Th2 immune responses, efficient Aβ42 antibody levels, and disappearance of antigen specific T cells. In-vivo TNFRSF4/25 antibody co-stimulation enhanced Aβ42 specific T cell responses with initial Th2 expansion and subsequent development of Aβ42 specific CD4+CD25+Foxp3+ cells. It showed that Th2 biased responses due to gene gun immunizations propagate the development of regulatory T cells. In conclusion, full-length DNA Aβ42 immunization into skin results in a regulatory response with minimal risk of inflammation and autoimmunity.
Collapse
Affiliation(s)
- Doris Lambracht-Washington
- Department of Neurology and Neurotherapeutics, Alzheimer's Disease Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Roger N Rosenberg
- Department of Neurology and Neurotherapeutics, Alzheimer's Disease Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
340
|
Feldman HH, Gauthier S, Chertkow H, Conn DK, Freedman M, Chris M. PROGRESS IN CLINICAL NEUROSCIENCES: Canadian Guidelines for the Development of Antidementia Therapies: A Conceptual Summary. Can J Neurol Sci 2014; 33:6-26. [PMID: 16583717 DOI: 10.1017/s0317167100004649] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The magnitude of the problems faced by an aging Canadian society has been clearly identified. Perhaps the single most important problem is the increasing incidence of dementia. Alzheimer's disease (AD) accounts for 50-60% of the dementias in later life within a spectrum of other contributing dementias. Regulatory approval has been given to Acetylcholinesterase inhibitors for the symptomatic treatment of mild to moderate AD, and conditional approval to memantine for the symptoms of moderate to severe AD. There has been no regulatory approval for the treatment of the degenerative dementias beyond AD. The very rapid progress in the past decade in biotechnology and in the molecular biology of the dementias is supporting a new generation of innovative treatment strategies that will more directly target the underlying disease pathogenic mechanisms. Such treatments will foreseeably include immunotherapies, anti-aggregants that may prevent misfolding and deposition of proteins, and neuroregenerative interventions. These Guidelines follow the 2nd Canadian Conference on the Development of Antidementia Therapies, held in 2004, which covered a range of design, methodological and ethical issues facing clinical researchers and regulatory authorities. They are intended to provide a common point of reference and guidance in Canada for therapeutic development of the dementias.
Collapse
Affiliation(s)
- Howard H Feldman
- Division of Neurology, University of British Columbia, Clinic for Alzheimer's Disease and Related Disorders, Vancouver, BC, Canada
| | | | | | | | | | | |
Collapse
|
341
|
Aβ immunotherapy for Alzheimer's disease: effects on apoE and cerebral vasculopathy. Acta Neuropathol 2014; 128:777-89. [PMID: 25195061 DOI: 10.1007/s00401-014-1340-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 08/28/2014] [Accepted: 08/30/2014] [Indexed: 12/30/2022]
Abstract
Aβ immunotherapy for Alzheimer's disease (AD) results in the removal of Aβ plaques and increased cerebral amyloid angiopathy (CAA). In current clinical trials, amyloid-related imaging abnormalities (ARIAs), putatively due to exacerbation of CAA, are concerning side effects. We aimed to assess the role of the Aβ transporter apolipoprotein E (apoE) in the exacerbation of CAA and development of CAA-associated vasculopathy after Aβ immunotherapy. 12 Aβ42-immunized AD (iAD; AN1792, Elan Pharmaceuticals) cases were compared with 28 unimmunized AD (cAD) cases. Immunohistochemistry was quantified for Aβ42, apoE, apoE E4 and smooth muscle actin, and CAA-associated vasculopathy was analyzed. Aβ immunotherapy was associated with redistribution of apoE from cortical plaques to cerebral vessel walls, mirroring the altered distribution of Aβ42. Concentric vessel wall splitting was increased threefold in leptomeningeal vessels after immunotherapy (cAD 6.3 vs iAD 20.6 %, P < 0.001), but smooth muscle cell abnormalities did not differ. The findings suggest that apoE is involved in the removal of plaques and transport of Aβ to the cerebral vasculature induced by Aβ immunotherapy. Immunotherapy was not associated with CAA-related vascular smooth muscle damage, but was accompanied by increased splitting of the vessel wall, perhaps reflecting enhanced deposition and subsequent removal of Aβ. ARIA occurring in some current trials of Aβ immunotherapy may reflect an extreme form of these vascular changes.
Collapse
|
342
|
Delrieu J, Ousset P, Voisin T, Vellas B. Amyloid beta peptide immunotherapy in Alzheimer disease. Rev Neurol (Paris) 2014; 170:739-48. [DOI: 10.1016/j.neurol.2014.10.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 09/11/2014] [Accepted: 10/03/2014] [Indexed: 11/28/2022]
|
343
|
Abstract
INTRODUCTION Activated microglia are associated with the progression of Alzheimer's disease (AD), as well as many other neurodegenerative diseases of aging. Microglia are therefore key targets for therapeutic intervention. AREAS COVERED β-amyloid (Aβ) deposits activate the complement system, which, in turn, stimulates microglia to release neurotoxic materials. Research has focused primarily on anti-inflammatory agents to temper this toxic effect. More recently there has been a focus on converting microglia from this M1 state to an M2 state in which the toxic effects are reduced and their phagocytic activity toward Aβ enhanced. Studies in transgenic mice have suggested a number of possible anti-inflammatory approaches but they may not always be a good model. An example is vaccination with antibodies to Aβ, which is effective in mouse models, but has repeatedly failed in clinical trials. Biomarker studies indicate that AD commences many years prior to clinical onset. EXPERT OPINION A hopeful approach to a disease-modifying treatment of AD is to administer agents that inhibit the inflammatory stimulation of microglia or successfully convert them to an M2 state. However, any such treatment must be started early in the disease.
Collapse
Affiliation(s)
- Patrick L McGeer
- University of British Columbia, Kinsmen Laboratory of Neurological Research , 2255 Wesbrook Mall, Vancouver, BC V6T 1Z3 , Canada
| | | |
Collapse
|
344
|
Peptide Vaccines for Hypertension and Diabetes Mellitus. Vaccines (Basel) 2014; 2:832-40. [PMID: 26344893 PMCID: PMC4494253 DOI: 10.3390/vaccines2040832] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 10/10/2014] [Accepted: 11/03/2014] [Indexed: 12/25/2022] Open
Abstract
Vaccines are commonly used as a preventive medicine for infectious diseases worldwide; however, the trial for an amyloid beta vaccine against Alzheimer’s disease will open a new concept in vaccination. In case of therapeutic vaccines for cancer, their targets are usually specific antigens in cancer cells, allowing activated cytotoxic T cells (CTLs) to attach and remove the antigen-presenting cancer cells. In our therapeutic vaccines against hypertension, the target is angiotensin II (Ang II) and induced anti-Ang II antibodies could efficiently ameliorate high blood pressure. Similarly, we developed the therapeutic vaccine against DPP4 for diabetes mellitus. However, because Ang II or DPP4 is an endogenous hormone, we must avoid autoimmune disease induced by these vaccines. Therefore, our system was used to design a therapeutic vaccine that elicits anti-Ang II or DPP4 antibodies without CTL activation against Ang II or DPP4. In this review, we will describe our concept of therapeutic vaccines for hypertension and diabetes mellitus.
Collapse
|
345
|
Zhang Y, Zhang H, Zhang G, Ka K, Huang W. Combining acellular nerve allografts with brain-derived neurotrophic factor transfected bone marrow mesenchymal stem cells restores sciatic nerve injury better than either intervention alone. Neural Regen Res 2014; 9:1814-9. [PMID: 25422643 PMCID: PMC4239771 DOI: 10.4103/1673-5374.143427] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2014] [Indexed: 01/08/2023] Open
Abstract
In this study, we chemically extracted acellular nerve allografts from bilateral sciatic nerves, and repaired 10-mm sciatic nerve defects in rats using these grafts and brain-derived neurotrophic factor transfected bone marrow mesenchymal stem cells. Experiments were performed in three groups: the acellular nerve allograft bridging group, acellular nerve allograft + bone marrow mesenchymal stem cells group, and the acellular nerve allograft + brain-derived neurotrophic factor transfected bone marrow mesenchymal stem cells group. Results showed that at 8 weeks after bridging, sciatic functional index, triceps wet weight recovery rate, myelin thickness, and number of myelinated nerve fibers were significantly changed in the three groups. Variations were the largest in the acellular nerve allograft + brain-derived neurotrophic factor transfected bone marrow mesenchymal stem cells group compared with the other two groups. Experimental findings suggest that chemically extracted acellular nerve allograft combined nerve factor and mesenchymal stem cells can promote the restoration of sciatic nerve defects. The repair effect seen is better than the single application of acellular nerve allograft or acellular nerve allograft combined mesenchymal stem cell transplantation.
Collapse
Affiliation(s)
- Yanru Zhang
- School of International Education, Zhengzhou University, Zhengzhou, Henan Province, China ; Institute of Clinical Anatomy, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Hui Zhang
- Department of Orthopedic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Gechen Zhang
- Department of Orthopedic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Ka Ka
- School of International Education, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Wenhua Huang
- Institute of Clinical Anatomy, Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
346
|
Anderson KM, Olson KE, Estes KA, Flanagan K, Gendelman HE, Mosley RL. Dual destructive and protective roles of adaptive immunity in neurodegenerative disorders. Transl Neurodegener 2014; 3:25. [PMID: 25671101 PMCID: PMC4323229 DOI: 10.1186/2047-9158-3-25] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 10/28/2014] [Indexed: 12/14/2022] Open
Abstract
Inappropriate T cell responses in the central nervous system (CNS) affect the pathogenesis of a broad range of neuroinflammatory and neurodegenerative disorders that include, but are not limited to, multiple sclerosis, amyotrophic lateral sclerosis, Alzheimer’s disease and Parkinson’s disease. On the one hand immune responses can exacerbate neurotoxic responses; while on the other hand, they can lead to neuroprotective outcomes. The temporal and spatial mechanisms by which these immune responses occur and are regulated in the setting of active disease have gained significant recent attention. Spatially, immune responses that affect neurodegeneration may occur within or outside the CNS. Migration of antigen-specific CD4+ T cells from the periphery to the CNS and consequent immune cell interactions with resident glial cells affect neuroinflammation and neuronal survival. The destructive or protective mechanisms of these interactions are linked to the relative numerical and functional dominance of effector or regulatory T cells. Temporally, immune responses at disease onset or during progression may exhibit a differential balance of immune responses in the periphery and within the CNS. Immune responses with predominate T cell subtypes may differentially manifest migratory, regulatory and effector functions when triggered by endogenous misfolded and aggregated proteins and cell-specific stimuli. The final result is altered glial and neuronal behaviors that influence the disease course. Thus, discovery of neurodestructive and neuroprotective immune mechanisms will permit potential new therapeutic pathways that affect neuronal survival and slow disease progression.
Collapse
Affiliation(s)
- Kristi M Anderson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, The University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Katherine E Olson
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, The University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Katherine A Estes
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, The University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Ken Flanagan
- Prothena Biosciences, South San Francisco, 650 Gateway Boulevard, CA 94080 USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, The University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - R Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, The University of Nebraska Medical Center, Omaha, NE 68198 USA
| |
Collapse
|
347
|
Guo W, Sha S, Jiang T, Xing X, Cao Y. A new DNA vaccine fused with the C3d-p28 induces a Th2 immune response against amyloid-beta. Neural Regen Res 2014; 8:2581-90. [PMID: 25206569 PMCID: PMC4145937 DOI: 10.3969/j.issn.1673-5374.2013.27.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Accepted: 08/02/2013] [Indexed: 01/11/2023] Open
Abstract
To enhance anti-amyloid-beta (Aβ) antibody generation and induce a Th2 immune response, we constructed a new DNA vaccine p(Aβ3–10)10-C3d-p28.3 encoding ten repeats of Aβ3–10 and three copies of C3d-p28 as a molecular adjuvant. In this study, we administered this adjuvant cularly to female C57BL/6J mice at 8–10 weeks of age. Enzyme linked immunosorbent assay was used to detect the titer of serum anti-Aβ antibody, isotypes, and cytokines in splenic T cells. A 3-(4,5-cimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay was used to detect the prolifera-tion rate of splenic T cells. Brain sections from a 12-month-old APP/PS1 transgenic mouse were used for detecting the binding capacities of anti-Aβ antibodies to Aβ plaques. The p(Aβ3–10)10-C3d-p28.3 vaccine induced high titers of anti-amyloid-β antibodies, which bound to Aβ plaques in APP/PS1 transgenic mouse brain tissue, demonstrating that the vaccine is effective against plaques in a mouse model of Alzheimer's disease. Moreover, the vaccine elicited a predo-minantly IgG1 humoral response and low levels of interferon-γ in ex vivo cultured splenocytes, dicating that the vaccine could shift the cellular immune response towards a Th2 phenotype. This indicated that the vaccine did not elicit a detrimental immune response and had a favorable safety profile. Our results indicate that the p(Aβ3–10)10-C3d-p28.3 vaccine is a promising immunothe-peutic option for Aβ vaccination in Alzheimer's disease.
Collapse
Affiliation(s)
- Wanshu Guo
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Sha Sha
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Tongzi Jiang
- Department of Neurology, First People's Hospital of Shenyang City, Shenyang 110041, Liaoning Province, China
| | - Xiaona Xing
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Yunpeng Cao
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| |
Collapse
|
348
|
Yuan Z, Du M, Chen Y, Dou F. Construction of human Fab library and screening of a single-domain antibody of amyloid-beta 42 oligomers. Neural Regen Res 2014; 8:3107-15. [PMID: 25206631 PMCID: PMC4158702 DOI: 10.3969/j.issn.1673-5374.2013.33.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 07/01/2013] [Indexed: 01/25/2023] Open
Abstract
Screening humanized antibodies from a human Fab phage display library is an effective and quick method to obtain beta-amyloid oligomers. Thus, the present study prepared amyloid-beta 42 oli-gomers and constructed a naïve human Fab phage display library based on blood samples from six healthy people. After three rounds of biopanning in vitro, a human single-domain antibody that specifically recognized amyloid-beta 42 oligomers was identified. Western blot and enzyme-linked immunosorbent assay demonstrated this antibody bound specifically to human amyloid-beta 42 tetramer and nonamer, but not the monomer or high molecular weight oligomers. This study successfully constructed a human phage display library and screened a single-domain antibody that specifically recognized amyloid-beta 42 oligomers.
Collapse
Affiliation(s)
- Zuanning Yuan
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, Beijing 100875, China ; Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Minge Du
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, Beijing 100875, China ; Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Yiwen Chen
- Geriatric Internal Department of Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Fei Dou
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Beijing Normal University, Beijing 100875, China ; Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, Southeast University, Nanjing 210009, Jiangsu Province, China
| |
Collapse
|
349
|
Jiang T, Guo W, Sha S, Xing X, Guo R, Cao Y. Nasal mucosal inhalation of amyloid-beta peptide 3-10 defective adenovirus attenuates cytotoxicity induced by beta-amyloid (1-42). Neural Regen Res 2014; 9:872-7. [PMID: 25206904 PMCID: PMC4146261 DOI: 10.4103/1673-5374.131605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2014] [Indexed: 11/06/2022] Open
Abstract
Three-month-old Alzheimer's disease model transgenic mice were immunized with Aβ1–42 Plp-Adenovirus [Ad]-X-CMV-(Aβ3–10)10-CpG [AdCpG-(Aβ3–10)10] or AdCpG virus fluid via nasal mucosal inhalation, respectively. ELISA analysis of serum showed Aβ42 antibody titers were significantly increased in mice immunized with Aβ1–42 and AdCpG-(Aβ3–10)10. Concanavalin A and AdCpG-(Aβ3–10)10 stimulation significantly increased the number of proliferating spleen cells cultured from AdCpG(Aβ3–10)10 and Aβ42 groups compared with the control group. In the AdCpG(Aβ3–10)10 group, levels of interleukin (IL)-4 and IL-10 were increased, while those of IL-2 and interferon-γ were decreased. In the Aβ42 group, levels of IL-4, IL-10, IL-2 and interferon-γ were all increased. Experimental findings indicate that AdCpG-(Aβ3–10)10 vaccine can produce strong T helper 2 (Th2) humoral immune responses in addition to the production of Aβ42 antibody. The cellular immunologic response was weak and avoided Aβ1–42-mediated cytotoxicity.
Collapse
Affiliation(s)
- Tongzi Jiang
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Wanshu Guo
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Sha Sha
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xiaona Xing
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Rong Guo
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yunpeng Cao
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
350
|
Schindowski C, Zimmermann J, Schindowski K. Intravenous immunoglobulin for the treatment of Alzheimer's disease: current evidence and considerations. Degener Neurol Neuromuscul Dis 2014; 4:121-130. [PMID: 32669906 PMCID: PMC7337175 DOI: 10.2147/dnnd.s51786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 04/23/2014] [Indexed: 11/23/2022] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative form of dementia with increasing incidence rates in most countries. AD is characterized by amyloid plaques and neurofibrillary tangles in the brains of AD individuals accompanied by global neuronal loss. The peptide amyloid-β (Aβ) aggregates to amyloid plaques in AD brains. As a result, many therapeutic approaches target Aβ. Human plasma and the plasma product intravenous immunoglobulin (IVIG) contain naturally-occurring anti-Aβ antibodies (Nabs-Aβ) that appear to reduce risks of developing AD. IVIG sequesters Aβ and thus interferes with AD progression. This study reviews the role of different Aβ species, Nabs-Aβ, preclinical data, and clinical studies of IVIG as potential AD treatments. The focus of this study is the outcomes of a recent Gammaglobulin Alzheimer's Partnership Phase III trial that did not reach primary endpoints, as well as efforts to compare IVIG with current anti-Aβ monoclonals such as bapineuzumab, solanezumab, and BIIB037. Moreover, this study critically examines current market and ethical consequences of potential off-label uses of IVIG, limits in IVIG supply, and subsequent challenges.
Collapse
Affiliation(s)
- Christina Schindowski
- Vivantes Klinikum am Urban Hospital, Department of Psychiatry, Psychotherapy, and Psychosomatic Medicine, Berlin, Germany
| | | | - Katharina Schindowski
- Institute of Applied Biotechnology, Faculty for Biotechnology, Biberach University of Applied Sciences, Biberach/Riss, Germany
| |
Collapse
|