101
|
Zeitlinger M, Bauer M, Reindl-Schwaighofer R, Stoekenbroek RM, Lambert G, Berger-Sieczkowski E, Lagler H, Oesterreicher Z, Wulkersdorfer B, Lührs P, Galabova G, Schwenke C, Mader RM, Medori R, Landlinger C, Kutzelnigg A, Staffler G. A phase I study assessing the safety, tolerability, immunogenicity, and low-density lipoprotein cholesterol-lowering activity of immunotherapeutics targeting PCSK9. Eur J Clin Pharmacol 2021; 77:1473-1484. [PMID: 33969434 PMCID: PMC8440313 DOI: 10.1007/s00228-021-03149-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/20/2021] [Indexed: 11/28/2022]
Abstract
Purpose AT04A and AT06A are two AFFITOPE® peptide vaccine candidates being developed for the treatment of hypercholesterolemia by inducing proprotein convertase subtilisin/kexin type 9 (PCSK9)-specific antibodies. This study aimed to investigate safety, tolerability, antibody development, and reduction of low-density lipoprotein cholesterol (LDLc) following four subcutaneous immunizations. Methods This phase I, single-blind, randomized, placebo-controlled study was conducted in a total of 72 healthy subjects with a mean fasting LDLc level at baseline of 117.1 mg/dL (range 77–196 mg/dL). Each cohort enrolled 24 subjects to receive three priming immunizations at weeks 0, 4, and 8 and to receive a single booster immunization at week 60 of either AT04A, AT06A, or placebo. In addition to safety (primary objective), the antigenic peptide- and PCSK9-specific antibody response and the impact on LDLc were evaluated over a period of 90 weeks. Results The most common systemic treatment-related adverse events (AEs) reported were fatigue, headache, and myalgia in 75% of subjects in the AT06A group and 58% and 46% of subjects in the placebo and AT04A groups, respectively. Injection site reactions (ISR) representing 63% of all treatment-emergent adverse events (TEAEs), were transient and mostly of mild or moderate intensity and rarely severe (3%). Both active treatments triggered a robust, long-lasting antibody response towards the antigenic peptides used for immunization that optimally cross-reacted with the target epitope on PCSK9. In the AT04A group, a reduction in serum LDLc was observed with a mean peak reduction of 11.2% and 13.3% from baseline compared to placebo at week 20 and 70 respectively, and over the whole study period, the mean LDLc reduction for the AT04A group vs. placebo was −7.2% (95% CI [−10.4 to −3.9], P < 0.0001). In this group, PCSK9 target epitope titers above 50 were associated with clinically relevant LDLc reductions with an individual maximal decrease of 39%. Conclusions Although both AT04A and AT06 were safe and immunogenic, only AT04A demonstrated significant LDLc-lowering activity, justifying further development. Trial registration EudraCT: 2015-001719-11. ClinicalTrials.gov
Identifier: NCT02508896. Supplementary Information The online version contains supplementary material available at 10.1007/s00228-021-03149-2.
Collapse
Affiliation(s)
- Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Martin Bauer
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Roman Reindl-Schwaighofer
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Robert M Stoekenbroek
- Department of Vascular Surgery, Academic Medical Center, University of Amsterdam, P.O. Box 22660, 1100 DD, Amsterdam, Netherlands
| | - Gilles Lambert
- Laboratoire Inserm, UMR 1188 DéTROI, Université de La Réunion, 2 Rue Maxime Rivière, 97490, Sainte Clotilde, France
| | - Evelyn Berger-Sieczkowski
- Department of Neurology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Heimo Lagler
- Department of Medicine I, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Zoe Oesterreicher
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Beatrix Wulkersdorfer
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Petra Lührs
- AFFiRiS AG, Karl Farkas Gasse 22, 1030, Vienna, Austria
| | - Gergana Galabova
- AFFiRiS AG, Karl Farkas Gasse 22, 1030, Vienna, Austria.,Origenis GmbH, Am Klopferspitz 19a, 82152, Martinsried, Germany
| | | | - Robert M Mader
- Department of Medicine I, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | | | | | | | | |
Collapse
|
102
|
Yakupova EI, Bobyleva LG, Shumeyko SA, Vikhlyantsev IM, Bobylev AG. Amyloids: The History of Toxicity and Functionality. BIOLOGY 2021; 10:biology10050394. [PMID: 34062910 PMCID: PMC8147320 DOI: 10.3390/biology10050394] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/26/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022]
Abstract
Proteins can perform their specific function due to their molecular structure. Partial or complete unfolding of the polypeptide chain may lead to the misfolding and aggregation of proteins in turn, resulting in the formation of different structures such as amyloid aggregates. Amyloids are rigid protein aggregates with the cross-β structure, resistant to most solvents and proteases. Because of their resistance to proteolysis, amyloid aggregates formed in the organism accumulate in tissues, promoting the development of various diseases called amyloidosis, for instance Alzheimer's diseases (AD). According to the main hypothesis, it is considered that the cause of AD is the formation and accumulation of amyloid plaques of Aβ. That is why Aβ-amyloid is the most studied representative of amyloids. Therefore, in this review, special attention is paid to the history of Aβ-amyloid toxicity. We note the main problems with anti-amyloid therapy and write about new views on amyloids that can play positive roles in the different organisms including humans.
Collapse
Affiliation(s)
- Elmira I. Yakupova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (L.G.B.); (S.A.S.); (I.M.V.); (A.G.B.)
- A. N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
- Correspondence: ; Tel.: +7-(985)687-77-27
| | - Liya G. Bobyleva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (L.G.B.); (S.A.S.); (I.M.V.); (A.G.B.)
| | - Sergey A. Shumeyko
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (L.G.B.); (S.A.S.); (I.M.V.); (A.G.B.)
| | - Ivan M. Vikhlyantsev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (L.G.B.); (S.A.S.); (I.M.V.); (A.G.B.)
| | - Alexander G. Bobylev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (L.G.B.); (S.A.S.); (I.M.V.); (A.G.B.)
| |
Collapse
|
103
|
Román GC, Gracia F, Torres A, Palacios A, Gracia K, Harris D. Acute Transverse Myelitis (ATM):Clinical Review of 43 Patients With COVID-19-Associated ATM and 3 Post-Vaccination ATM Serious Adverse Events With the ChAdOx1 nCoV-19 Vaccine (AZD1222). Front Immunol 2021; 12:653786. [PMID: 33981305 PMCID: PMC8107358 DOI: 10.3389/fimmu.2021.653786] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/08/2021] [Indexed: 01/13/2023] Open
Abstract
Introduction Although acute transverse myelitis (ATM) is a rare neurological condition (1.34-4.6 cases per million/year) COVID-19-associated ATM cases have occurred during the pandemic. Case-finding methods We report a patient from Panama with SARS-CoV-2 infection complicated by ATM and present a comprehensive clinical review of 43 patients with COVID-19-associated ATM from 21 countries published from March 2020 to January 2021. In addition, 3 cases of ATM were reported as serious adverse events during the clinical trials of the COVID-19 vaccine ChAdOx1 nCoV-19 (AZD1222). Results All patients had typical features of ATM with acute onset of paralysis, sensory level and sphincter deficits due to spinal cord lesions demonstrated by imaging. There were 23 males (53%) and 20 females (47%) ranging from ages 21- to 73- years-old (mean age, 49 years), with two peaks at 29 and 58 years, excluding 3 pediatric cases. The main clinical manifestations were quadriplegia (58%) and paraplegia (42%). MRI reports were available in 40 patients; localized ATM lesions affected ≤3 cord segments (12 cases, 30%) at cervical (5 cases) and thoracic cord levels (7 cases); 28 cases (70%) had longitudinally-extensive ATM (LEATM) involving ≥4 spinal cord segments (cervicothoracic in 18 cases and thoracolumbar-sacral in 10 patients). Acute disseminated encephalomyelitis (ADEM) occurred in 8 patients, mainly women (67%) ranging from 27- to 64-years-old. Three ATM patients also had blindness from myeloneuritis optica (MNO) and two more also had acute motor axonal neuropathy (AMAN). Conclusions We found ATM to be an unexpectedly frequent neurological complication of COVID-19. Most cases (68%) had a latency of 10 days to 6 weeks that may indicate post-infectious neurological complications mediated by the host’s response to the virus. In 32% a brief latency (15 hours to 5 days) suggested a direct neurotropic effect of SARS-CoV-2. The occurrence of 3 reported ATM adverse effects among 11,636 participants in the AZD1222 vaccine trials is extremely high considering a worldwide incidence of 0.5/million COVID-19-associated ATM cases found in this report. The pathogenesis of ATM remains unknown, but it is conceivable that SARS-CoV-2 antigens –perhaps also present in the AZD1222 COVID-19 vaccine or its chimpanzee adenovirus adjuvant– may induce immune mechanisms leading to the myelitis.
Collapse
Affiliation(s)
- Gustavo C Román
- Department of Neurology, Neurological Institute, Houston Methodist Hospital, Houston, TX, United States.,Weill Cornell College of Medicine, Cornell University, New York, NY, United States.,Department of Neurology, Texas A&M University College of Medicine, Bryan, TX, United States
| | - Fernando Gracia
- Neurology Service, Hospital Paitilla, Panama City, Panama.,Faculty of Health Sciences, Interamerican University of Panama, Panama City, Panama.,Neurology Service, Hospital Santo Tomás, Panama City, Panama
| | - Antonio Torres
- Infectious Disease Service, Hospital Santo Tomás, Panama City, Panama
| | - Alexis Palacios
- Neuroradiology Service, Complejo Hospitalario Metropolitano, CSS (Caja de Seguro Social), Panama City, Panama
| | - Karla Gracia
- Interamerican University of Panama, Panama City, Panama
| | - Diógenes Harris
- Neurosurgery Service, Complejo Hospitalario Metropolitano, CSS, Panama City, Panama
| |
Collapse
|
104
|
Albus A, Kronimus Y, Neumann S, Vidovic N, Frenzel A, Kuhn P, Seifert M, Ziehm T, van der Wurp H, Dodel R. Effects of a Multimerized Recombinant Autoantibody Against Amyloid-β. Neuroscience 2021; 463:355-369. [PMID: 33958140 DOI: 10.1016/j.neuroscience.2021.03.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease; thus, the search for a cure or causal therapy has become necessary. Despite intense research on this topic in recent decades, there is no curative therapy up today, and also no disease-modifying treatment has been approved. As promising approach passive immunization strategies have thereby come forth. In this study, we focused on naturally occurring autoantibodies against the AD-associated peptide amyloid-β. These antibodies have already reported to show beneficial functions in vitro and in mouse models of AD. However, their availability is limited due to their low abundance in peripheral blood. In a recent study, we were able to generate four recombinant antibodies against amyloid-β. In the present study, we tested these antibodies in ELISA and SPR assays for their binding behavior and by aggregation- and phagocytosis assays as functional evidences to characterize their amyloid-β-related neutralizing and clearance abilities. Further ex vivo assay on organotypic hippocampal slice cultures gave first evidence of microglial activation and inflammatory features. The tested recombinant antibodies in IgG format showed, in comparison to naturally occurring autoantibodies against amyloid-β, insufficient binding capacities and -affinities. However, after conversion of one antibody into a single chain format multimerization of the scFv-Fc construct, the investigated binding capacity and -affinity showed improvements. Further functional assays predict a protective effect of this antibody. Although, all four recombinant antibodies showed binding to amyloid-β, promising features were only detectable after conversion into a multimeric format. The multimeric scFv-Fc antibody exhibited thereby strong impact on amyloid-β clearance and inhibition of oligomerization.
Collapse
Affiliation(s)
- Alexandra Albus
- Chair of Geriatric Medicine, University Duisburg-Essen, Germany; Department of Neurology, Philipps-University, Marburg, Germany
| | - Yannick Kronimus
- Chair of Geriatric Medicine, University Duisburg-Essen, Germany; Department of Neurology, Philipps-University, Marburg, Germany
| | - Sascha Neumann
- Chair of Geriatric Medicine, University Duisburg-Essen, Germany
| | - Natascha Vidovic
- Chair of Geriatric Medicine, University Duisburg-Essen, Germany; Department of Neurology, Philipps-University, Marburg, Germany
| | | | | | - Marc Seifert
- Institute of Cell Biology (Cancer Research), Medical Faculty, University Duisburg-Essen, Germany
| | - Tamar Ziehm
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich, Germany
| | - Hendrik van der Wurp
- Chair of Geriatric Medicine, University Duisburg-Essen, Germany; Faculty of Statistics, TU Dortmund University, Dortmund, Germany
| | - Richard Dodel
- Chair of Geriatric Medicine, University Duisburg-Essen, Germany; Department of Neurology, Philipps-University, Marburg, Germany.
| |
Collapse
|
105
|
Chwalisz BK. Cerebral amyloid angiopathy and related inflammatory disorders. J Neurol Sci 2021; 424:117425. [PMID: 33840507 DOI: 10.1016/j.jns.2021.117425] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/27/2020] [Accepted: 03/24/2021] [Indexed: 11/19/2022]
Abstract
Inflammatory cerebral amyloid angiopathy is a largely reversible inflammatory vasculopathy that develops in an acute or subacute fashion in reaction to amyloid protein deposition in the central nervous system blood vessels. There are two recognized pathologically characterized variants: cerebral amyloid angiopathy-related inflammation (CAAri) and A beta-related angiitis (ABRA). Both variants produce a clinical picture that resembles primary angiitis of the CNS but is distinguished by a characteristic radiologic appearance. Although originally defined as a clinicopathologic diagnosis, it can now often be diagnosed based on clinicoradiologic criteria, though confirmation with brain and meningeal biopsy is still required in some cases. This disorder typically responds to steroids but addition of other immune suppressants may be needed in some cases to control the disease.
Collapse
Affiliation(s)
- B K Chwalisz
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, 15 Parkman Street, Suite 835, Boston, MA 02114, USA; Division of Neuro-Ophthalmology, Department of Ophthalmology, Massachusetts Eye & Ear Infirmary/Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
106
|
Cercy SP. Pericytes and the Neurovascular Unit: The Critical Nexus of Alzheimer Disease Pathogenesis? EXPLORATORY RESEARCH AND HYPOTHESIS IN MEDICINE 2021. [DOI: 10.14218/erhm.2020.00062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
107
|
Reiss AB, Montufar N, DeLeon J, Pinkhasov A, Gomolin IH, Glass AD, Arain HA, Stecker MM. Alzheimer Disease Clinical Trials Targeting Amyloid: Lessons Learned From Success in Mice and Failure in Humans. Neurologist 2021; 26:52-61. [PMID: 33646990 DOI: 10.1097/nrl.0000000000000320] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND The goal of slowing or halting the development of Alzheimer disease (AD) has resulted in the huge allocation of resources by academic institutions and pharmaceutical companies to the development of new treatments. The etiology of AD is elusive, but the aggregation of amyloid-β and tau peptide and oxidative processes are considered critical pathologic mechanisms. The failure of drugs with multiple mechanisms to meet efficacy outcomes has caused several companies to decide not to pursue further AD studies and has left the field essentially where it has been for the past 15 years. Efforts are underway to develop biomarkers for detection and monitoring of AD using genetic, imaging, and biochemical technology, but this is of minimal use if no intervention can be offered. REVIEW SUMMARY In this review, we consider the natural progression of AD and how it continues despite present attempts to modify the amyloid-related machinery to alter the disease trajectory. We describe the mechanisms and approaches to AD treatment targeting amyloid, including both passive and active immunotherapy as well as inhibitors of enzymes in the amyloidogenic pathway. CONCLUSION Lessons learned from clinical trials of amyloid reduction strategies may prove crucial for the leap forward toward novel therapeutic targets to treat AD.
Collapse
Affiliation(s)
- Allison B Reiss
- Department of Medicine, NYU Long Island School of Medicine, Mineola, NY
| | - Natalie Montufar
- Department of Medicine, NYU Long Island School of Medicine, Mineola, NY
| | - Joshua DeLeon
- Department of Medicine, NYU Long Island School of Medicine, Mineola, NY
| | - Aaron Pinkhasov
- Department of Medicine, NYU Long Island School of Medicine, Mineola, NY
| | - Irving H Gomolin
- Department of Medicine, NYU Long Island School of Medicine, Mineola, NY
| | - Amy D Glass
- Department of Medicine, NYU Long Island School of Medicine, Mineola, NY
| | - Hirra A Arain
- Department of Medicine, NYU Long Island School of Medicine, Mineola, NY
| | - Mark M Stecker
- Fresno Center for Medical Education and Research, Department of Medicine, University of California-San Francisco, Fresno, CA
| |
Collapse
|
108
|
Relapsing polychondritis coupling with cerebral amyloid deposit inducing cerebral amyloid angiopathy-related inflammation. J Thromb Thrombolysis 2021; 49:681-684. [PMID: 32253708 DOI: 10.1007/s11239-020-02109-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Cerebral amyloid angiopathy-related inflammation is a syndrome of reversible encephalopathy with cerebral amyloid angiopathy, however the pathology is not well understood. We clear a part of the pathology through the first case of an 80-year-old man with cerebral amyloid angiopathy-related inflammation induced by relapsing polychondritis (RP) analysis. An 80-year-old man was diagnosed with RP by auricular cartilage biopsy. Almost no abnormality including intracranial microbleeding was detected by cranial magnetic resonance image (MRI) at diagnosis. However, he developed a headache and hallucination after five months. Seven-month cranial MRI showed novel, multiple, intracranial microbleeding, especially in the bilateral but asymmetry posterior, temporal, and parietal lobes. 123I-N-isopropyl-p-iodoamphetamine single-photon emission computed tomography showed increased cerebral blood flow in the bilateral posterior lobes. After treatment, both of his neurological symptoms and increased cerebral blood flow improved to mild. Photon emission computed tomography using Pittsburgh compound B (PiB) for evaluation of brain amyloidosis at 12 months after onset showed an amyloid deposit in the bilateral frontal lobes, but a lack of uptake corresponded to the RP lesions. Our case suggests that inflammation coupled with an amyloid deposit, induced the multiple intracranial bleeding, and resulted in the lack of PiB uptake. Findings from our case show that inflammation including excess blood flow coupled with an amyloid deposit synergistically facilitate intracranial bleeding.
Collapse
|
109
|
Shi M, Chu F, Tian X, Aerqin Q, Zhu F, Zhu J. Role of Adaptive Immune and Impacts of Risk Factors on Adaptive Immune in Alzheimer's Disease: Are Immunotherapies Effective or Off-Target? Neuroscientist 2021; 28:254-270. [PMID: 33530843 DOI: 10.1177/1073858420987224] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The pathogenesis of Alzheimer's disease (AD) is complex. Still it remains unclear, which resulted in all efforts for AD treatments with targeting the pathogenic factors unsuccessful over past decades. It has been evidenced that the innate immune is strongly implicated in the pathogenesis of AD. However, the role of adaptive immune in AD remains mostly unknown and the results obtained were controversial. In the review, we summarized recent studies and showed that the molecular and cellular alterations in AD patients and its animal models involving T cells and B cells as well as immune mediators of adaptive immune occur not only in the peripheral blood but also in the brain and the cerebrospinal fluid. The risk factors that cause AD contribute to AD progress by affecting the adaptive immune, indicating that adaptive immunity proposes a pivotal role in this disease. It may provide a possible basis for applying immunotherapy in AD and further investigates whether the immunotherapies are effective or off-target?
Collapse
Affiliation(s)
- Mingchao Shi
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China.,Department of Neurobiology, Care Sciences & Society, Division of Neurogeriatrcs, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Fengna Chu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China.,Department of Neurobiology, Care Sciences & Society, Division of Neurogeriatrcs, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Xiaoping Tian
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital of Shenzhen University Medical College, Shenzhen, China
| | - Qiaolifan Aerqin
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Feiqi Zhu
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital of Shenzhen University Medical College, Shenzhen, China
| | - Jie Zhu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China.,Department of Neurobiology, Care Sciences & Society, Division of Neurogeriatrcs, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| |
Collapse
|
110
|
Abstract
The history of Alzheimer's disease (AD) started in 1907, but we needed to wait until the end of the century to identify the components of pathological hallmarks and genetic subtypes and to formulate the first pathogenic hypothesis. Thanks to biomarkers and new technologies, the concept of AD then rapidly changed from a static view of an amnestic dementia of the presenium to a biological entity that could be clinically manifested as normal cognition or dementia of different types. What is clearly emerging from studies is that AD is heterogeneous in each aspect, such as amyloid composition, tau distribution, relation between amyloid and tau, clinical symptoms, and genetic background, and thus it is probably impossible to explain AD with a single pathological process. The scientific approach to AD suffers from chronological mismatches between clinical, pathological, and technological data, causing difficulty in conceiving diagnostic gold standards and in creating models for drug discovery and screening. A recent mathematical computer-based approach offers the opportunity to study AD in real life and to provide a new point of view and the final missing pieces of the AD puzzle.
Collapse
Affiliation(s)
- Camilla Ferrari
- Department of Neuroscience, Psychology, Drug Research, and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research, and Child Health (NEUROFARBA), University of Florence, Florence, Italy.,IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| |
Collapse
|
111
|
Lambracht-Washington D, Fu M, Hynan LS, Rosenberg RN. Changes in the brain transcriptome after DNA Aβ42 trimer immunization in a 3xTg-AD mouse model. Neurobiol Dis 2021; 148:105221. [PMID: 33316368 PMCID: PMC7845550 DOI: 10.1016/j.nbd.2020.105221] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 11/23/2020] [Accepted: 12/07/2020] [Indexed: 12/23/2022] Open
Abstract
Alzheimer's disease (AD) has been associated with accumulation of amyloid beta (Aβ) peptides in brain, and immunotherapy targeting Aβ provides potential for AD prevention. We have used a DNA Aβ42 trimer construct for immunization of 3xTg-AD mice and found previously significant reduction of amyloid and tau pathology due to the immunotherapy. We show here that DNA Aβ42 immunized 3xTg-AD mice showed better performance in nest building activities and had a higher 24 months survival rate compared to the non-treated AD controls. The analysis of differently expressed genes in brains from 24 months old mice showed significant increases transcript levels between non-immunized AD mice and wild-type controls for genes involved in microglia and astrocyte function, cytokine and inflammatory signaling, apoptosis, the innate and adaptive immune response and are consistent with an inflammatory phenotype in AD. Most of these upregulated genes were downregulated in the DNA Aβ42 immunized 3xTg-AD mice due to the vaccine. Transcript numbers for the immediate early genes, Arc, Bdnf, Homer1, Egr1 and cfos, involved in neuronal and neurotransmission pathways which were much lower in the non-immunized 3xTg-AD mice, were restored to wild-type mouse brain levels in DNA Aβ42 immunized 3xTg-AD mice indicating positive effects of DNA Aβ42 immunotherapy on synapse stability and plasticity. The immune response after immunization is complex, but the multitude of changes after DNA Aβ42 immunization shows that this response moves beyond the amyloid hypothesis and into direction of disease prevention.
Collapse
Affiliation(s)
- Doris Lambracht-Washington
- Department of Neurology, UT Southwestern Medical Center Dallas, USA; Doris Lambracht Washington, UT Southwestern Medical Center Dallas, Department of Neurology , 5323 Harry Hines Blvd, Dallas, TX 75390-8813, USA.
| | - Min Fu
- Department of Neurology, UT Southwestern Medical Center Dallas, USA.
| | - Linda S Hynan
- Departments of Population and Data Sciences (Biostatistics) & Psychiatry, UT Southwestern Medical Center Dallas, USA.
| | - Roger N Rosenberg
- Department of Neurology, UT Southwestern Medical Center Dallas, USA.
| |
Collapse
|
112
|
[The future of dementia prevention and treatment strategies]. Nihon Ronen Igakkai Zasshi 2020; 57:374-396. [PMID: 33268621 DOI: 10.3143/geriatrics.57.374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
113
|
Mantile F, Prisco A. Vaccination against β-Amyloid as a Strategy for the Prevention of Alzheimer's Disease. BIOLOGY 2020; 9:425. [PMID: 33260956 PMCID: PMC7761159 DOI: 10.3390/biology9120425] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 12/17/2022]
Abstract
Vaccination relies on the phenomenon of immunity, a long-term change in the immunological response to subsequent encounters with the same pathogen that occurs after the recovery from some infectious diseases. However, vaccination is a strategy that can, in principle, be applied also to non-infectious diseases, such as cancer or neurodegenerative diseases, if an adaptive immune response can prevent the onset of the disease or modify its course. Immunization against β-amyloid has been explored as a vaccination strategy for Alzheimer's disease for over 20 years. No vaccine has been licensed so far, and immunotherapy has come under considerable criticism following the negative results of several phase III clinical trials. In this narrative review, we illustrate the working hypothesis behind immunization against β-amyloid as a vaccination strategy for Alzheimer's disease, and the outcome of the active immunization strategies that have been tested in humans. On the basis of the lessons learned from preclinical and clinical research, we discuss roadblocks and current perspectives in this challenging enterprise in translational immunology.
Collapse
|
114
|
Alzheimer's disease: Recent treatment strategies. Eur J Pharmacol 2020; 887:173554. [DOI: 10.1016/j.ejphar.2020.173554] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/03/2020] [Accepted: 09/11/2020] [Indexed: 12/12/2022]
|
115
|
Weninger S, Sperling B, Alexander R, Ivarsson M, Menzies FM, Powchik P, Weber CJ, Altar CA, Crystal RG, Haggarty SJ, Loring J, Bain LJ, Carrillo MC. Active immunotherapy and alternative therapeutic modalities for Alzheimer's disease. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2020; 6:e12090. [PMID: 33083513 PMCID: PMC7550557 DOI: 10.1002/trc2.12090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 09/11/2020] [Indexed: 12/17/2022]
Abstract
As knowledge of Alzheimer's disease (AD) progression improves, the field has recognized the need to diversify the pipeline, broaden strategies and approaches to therapies, as well as delivery mechanisms. A better understanding of the earliest biological processes of AD/dementia would help inform drug target selection. Currently there are a number of programs exploring these alternate avenues. This meeting will allow experts in the field (academia, industry, government) to provide perspectives and experiences that can help elucidate what the pipeline looks like today and what avenues hold promise in developing new therapies across the stages of AD. The focus here is on Active Immunotherapies and Alternative Therapeutic Modalities. This topic includes active vaccines, antisense oligomers, and cell-based therapy among others, and highlights new clinical developments that utilize these modalities.
Collapse
Affiliation(s)
| | | | - Robert Alexander
- Takeda Pharmaceuticals International Co. Cambridge Massachusetts USA
| | - Magnus Ivarsson
- Rodin Therapeutics 300 Technology Square Cambridge Massachusetts USA
| | | | - Peter Powchik
- United Neuroscience 9 Exchange Place, I. F. S. C Dublin Ireland
| | | | | | - Ronald G Crystal
- Department of Genetic Medicine Weill Cornell Medicine New York New York USA
| | - Stephen J Haggarty
- Chemical Neurobiology Laboratory Center for Genomic Medicine Department of Neurology Massachusetts General Hospital and Harvard Medical School Boston Massachusetts USA
| | | | - Lisa J Bain
- Independent Science Writer Elverson Pennsylvania USA
| | | |
Collapse
|
116
|
Wang L, Liu S, Xu J, Watanabe N, Mayo KH, Li J, Li X. Emodin inhibits aggregation of amyloid-β peptide 1-42 and improves cognitive deficits in Alzheimer's disease transgenic mice. J Neurochem 2020; 157:1992-2007. [PMID: 32799401 DOI: 10.1111/jnc.15156] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/09/2020] [Accepted: 08/11/2020] [Indexed: 12/22/2022]
Abstract
Aggregation of amyloid-β peptide 1-42 (Aβ42) initiates the onset of Alzheimer's disease (AD), and all the drugs designed to attenuate AD have failed in clinical trials. Emodin reduces levels of β-amyloid, tau aggregation, oxidative stress, and inflammatory response, demonstrating AD therapeutic potential, whereas its effect on the accumulation of the amyloid-β protein is not well understood. In this work, we investigated emodin activity on Aβ aggregation using a range of biochemical, biophysical, and cell-based approaches. We provide evidence to suggest that emodin blocks Aβ42 fibrillogenesis and Aβ-induced cytotoxicity, displaying a greater effect than that of curcumin. Through adopting three short peptides (Aβ1-16, Aβ17-33, and Aβ28-42), it was proven that emodin interacts with the Leu17-Gly33 sequence. Furthermore, our findings indicated that Val18 and Phe19 in Aβ42 are the target residues with which emodin interacts according amino acid mutation experiments. When fed to 8-month-old B6C3-Tg mice for 2 months, high-dose emodin ameliorates cognitive impairment by 60%-70%. Pathological results revealed that levels of Aβ deposition in the brains of AD mice treated with a high dose of emodin decreased by 50%-70%. Therefore, our study indicates that emodin may represent a promising drug for AD treatment.
Collapse
Affiliation(s)
- Lichun Wang
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| | - Sitong Liu
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal University, Changchun, China.,College of Life Sciences, Jilin University, Changchun, China
| | - Jiaqi Xu
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| | - Nobumoto Watanabe
- Bio-Active Compounds Discovery Research Unit, RIKEN Center for Sustainable Resource Science, Wako, Saitama, Japan
| | - Kevin H Mayo
- Biochemistry, Molecular Biology, and Biophysics, college of Biological Science, University of Minnesota, Minneapolis, MN, USA
| | - Jiang Li
- Affiliated Stomatology Hospital of Guangzhong Medical University, Guangzhou, China
| | - Xiaomeng Li
- The Key Laboratory of Molecular Epigenetics of MOE, Institute of Genetics and Cytology, Northeast Normal University, Changchun, China
| |
Collapse
|
117
|
Behl T, Kaur I, Fratila O, Brata R, Bungau S. Exploring the Potential of Therapeutic Agents Targeted towards Mitigating the Events Associated with Amyloid-β Cascade in Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21207443. [PMID: 33050199 PMCID: PMC7589257 DOI: 10.3390/ijms21207443] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/02/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023] Open
Abstract
One of the most commonly occurring neurodegenerative disorders, Alzheimer's disease (AD), encompasses the loss of cognitive and memory potential, impaired learning, dementia and behavioral defects, and has been prevalent since the 1900s. The accelerating occurrence of AD is expected to reach 65.7 million by 2030. The disease results in neural atrophy and disrupted inter-neuronal connections. Amongst multiple AD pathogenesis hypotheses, the amyloid beta (Aβ) cascade is the most relevant and accepted form of the hypothesis, which suggests that Aβ monomers are formed as a result of the cleavage of amyloid precursor protein (APP), followed by the conversion of these monomers to toxic oligomers, which in turn develop β-sheets, fibrils and plaques. The review targets the events in the amyloid hypothesis and elaborates suitable therapeutic agents that function by hindering the steps of plaque formation and lowering Aβ levels in the brain. The authors discuss treatment possibilities, including the inhibition of β- and γ-secretase-mediated enzymatic cleavage of APP, the immune response generating active immunotherapy and passive immunotherapeutic approaches targeting monoclonal antibodies towards Aβ aggregates, the removal of amyloid aggregates by the activation of enzymatic pathways or the regulation of Aβ circulation, glucagon-like peptide-1 (GLP-1)-mediated curbed accumulation and the neurotoxic potential of Aβ aggregates, bapineuzumab-mediated vascular permeability alterations, statin-mediated Aβ peptide degradation, the potential role of ibuprofen and the significance of natural drugs and dyes in hindering the amyloid cascade events. Thus, the authors aim to highlight the treatment perspective, targeting the amyloid hypothesis, while simultaneously emphasizing the need to conduct further investigations, in order to provide an opportunity to neurologists to develop novel and reliable treatment therapies for the retardation of AD progression.
Collapse
Affiliation(s)
- Tapan Behl
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India;
- Correspondence: (T.B.); (S.B.); Tel.: +40-726-776-588 (S.B.)
| | - Ishnoor Kaur
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India;
| | - Ovidiu Fratila
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, Oradea 410073, Romania; (O.F.); (R.B.)
| | - Roxana Brata
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, Oradea 410073, Romania; (O.F.); (R.B.)
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea 410028, Romania
- Correspondence: (T.B.); (S.B.); Tel.: +40-726-776-588 (S.B.)
| |
Collapse
|
118
|
Kawarabayashi T, Terakawa T, Takahashi A, Hasegawa H, Narita S, Sato K, Nakamura T, Seino Y, Hirohata M, Baba N, Ueda T, Harigaya Y, Kametani F, Maruyama N, Ishimoto M, St George-Hyslop P, Shoji M. Oral Immunization with Soybean Storage Protein Containing Amyloid-β 4-10 Prevents Spatial Learning Decline. J Alzheimers Dis 2020; 70:487-503. [PMID: 31177217 PMCID: PMC6700641 DOI: 10.3233/jad-190023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Amyloid-β (Aβ) plays a central role in the pathogenesis of Alzheimer’s disease (AD). Because AD pathologies begin two decades before the onset of dementia, prevention of Aβ amyloidosis has been proposed as a mean to block the pathological cascade. Here, we generate a transgenic plant-based vaccine, a soybean storage protein containing Aβ4–10, named Aβ+, for oral Aβ immunization. One mg of Aβ+ or control protein (Aβ–) was administered to TgCRND8 mice once a week from 9 weeks up to 58 weeks. Aβ+ immunization raised both anti-Aβ antibodies and cellular immune responses. Spatial learning decline was prevented in the Aβ+ immunized group in an extended reference memory version of Morris water maze test from 21 to 57 weeks. In Tris-buffered saline (TBS), sodium dodecyl sulfate (SDS), and formic acid (FA) serial extractions, all sets of Aβ species from Aβ monomer, low to high molecular weight Aβ oligomers, and Aβ smears had different solubility in TgCRND8 brains. Aβ oligomers decreased in TBS fractions, corresponding to an increase in high molecular weight Aβ oligomers in SDS extracts and Aβ smears in FA fraction of the Aβ+ treated group. There was significant inhibition of histological Aβ burden, especially in diffuse plaques, and suppression of microglial inflammation. Processing of amyloid-β protein precursor was not different between Aβ+ and Aβ– groups. No evidence of amyloid-related inflammatory angiopathy was observed. Thus, Aβ+ oral immunization could be a promising, cheap, and long-term safe disease-modifying therapy to prevent the pathological process in AD.
Collapse
Affiliation(s)
- Takeshi Kawarabayashi
- Department of Neurology, Geriatrics Research Institute Hospital, Maebashi, Aomori, Japan.,Department of Neurology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Teruhiko Terakawa
- Hokko Chemical Industry Co., Ltd, Atsugi-shi, Kanagawa, Japan.,Inplanta Innovations Inc. Yokohama, Kanagawa, Japan
| | | | | | - Sakiko Narita
- Department of Neurology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Kaoru Sato
- Department of Neurology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Takumi Nakamura
- Department of Neurology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan.,Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Yusuke Seino
- Department of Neurology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Mie Hirohata
- Department of Neurology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Nobue Baba
- Bioanalysis Department, LSI Medience Corporation, Itabashi-ku, Tokyo, Japan
| | - Tetsuya Ueda
- Bioanalysis Department, LSI Medience Corporation, Itabashi-ku, Tokyo, Japan
| | - Yasuo Harigaya
- Department of Neurology, Maebashi Red Cross Hospital, Maebashi, Japan
| | - Fuyuki Kametani
- Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | | | - Masao Ishimoto
- Institute of Crop Science, NARO, Tsukuba, Ibaraki, Japan
| | - Peter St George-Hyslop
- Tanz Centre for Research in Neurodegenerative Diseases, and Departments of Medicine, Medical Biophysics and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Mikio Shoji
- Department of Neurology, Geriatrics Research Institute Hospital, Maebashi, Aomori, Japan.,Department of Neurology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| |
Collapse
|
119
|
Poon CH, Wang Y, Fung ML, Zhang C, Lim LW. Rodent Models of Amyloid-Beta Feature of Alzheimer's Disease: Development and Potential Treatment Implications. Aging Dis 2020; 11:1235-1259. [PMID: 33014535 PMCID: PMC7505263 DOI: 10.14336/ad.2019.1026] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 10/26/2019] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder worldwide and causes severe financial and social burdens. Despite much research on the pathogenesis of AD, the neuropathological mechanisms remain obscure and current treatments have proven ineffective. In the past decades, transgenic rodent models have been used to try to unravel this disease, which is crucial for early diagnosis and the assessment of disease-modifying compounds. In this review, we focus on transgenic rodent models used to study amyloid-beta pathology in AD. We also discuss their possible use as promising tools for AD research. There is still no effective treatment for AD and the development of potent therapeutics are urgently needed. Many molecular pathways are susceptible to AD, ranging from neuroinflammation, immune response, and neuroplasticity to neurotrophic factors. Studying these pathways may shed light on AD pathophysiology as well as provide potential targets for the development of more effective treatments. This review discusses the advantages and limitations of these models and their potential therapeutic implications for AD.
Collapse
Affiliation(s)
- Chi Him Poon
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yingyi Wang
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Man-Lung Fung
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chengfei Zhang
- 2Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Lee Wei Lim
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
120
|
Dai M, Feng X, Mo Z, Sun Y, Fu L, Zhang Y, Wu J, Yu B, Zhang H, Yu X, Wu H, Kong W. Stimulation Effects and Mechanisms of Different Adjuvants on a Norovirus P Particle-Based Active Amyloid-β Vaccine. J Alzheimers Dis 2020; 77:1717-1732. [PMID: 32925038 DOI: 10.3233/jad-200351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Adjuvants are important components of vaccines and effectively enhance the immune response of specific antigens. However, the role of adjuvants or combinations of adjuvants in stimulating immunogenicity of the amyloid-β (Aβ) vaccine, as well as molecular mechanisms underlying such stimulation still remain unclear. A previous study of ours developed a norovirus P particle-based active Aβ epitope vaccine, PP-3copy-Aβ1-6-loop123, which stimulates a high titer of Aβ-specific antibodies in mouse Alzheimer's disease (AD) models. OBJECTIVE The most effective and safe adjuvant that maximizes the immunogenicity of our protein vaccine was determined. METHODS We investigated four adjuvants (CpG, AS02, AS03, and MF59), and combinations of those, for capacity to enhance immunogenicity, and performed transcriptome analysis to explore mechanisms underlying the role of these in AD immunotherapy. RESULTS Addition of the adjuvant, AS02, remarkably improved the immunogenicity of the PP-3copy-Aβ1-6-loop123 vaccine without triggering an Aβ-specific T-cell response. Combinations of adjuvants, particularly CpG + AS02 and CpG + AS03, elicited a significantly elevated and prolonged Aβ-specific antibody response. Gene ontology and Kyoto Encyclopedia of Genes and Genomes analyses indicated that a combination of two adjuvants was more effective in activating immune-related pathways, thereby enhancing the immunogenicity of PP-3copy-Aβ1-6-loop123. CONCLUSION These findings demonstrated that adjuvants can be used as enhancers in AD protein vaccination, and that a combination of CpG and AS-related adjuvants may be a very effective adjuvant candidate suitable for further clinical trials of the PP-3copy-Aβ1-6-loop123 vaccine. Our studies also revealed potential mechanisms underlying the stimulation of immune response of protein vaccines by adjuvants.
Collapse
Affiliation(s)
- MingRui Dai
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - XueJian Feng
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - ZengShuo Mo
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Yao Sun
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China
| | - Lu Fu
- Laboratory of Pathogenic Microbiology and Immunology, College of Life science, Jilin Agricultural University, Changchun, China
| | - Yong Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Jiaxin Wu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Bin Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Haihong Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Xianghui Yu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Hui Wu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, China.,Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| |
Collapse
|
121
|
Wang ZX, Wan Q, Xing A. HLA in Alzheimer's Disease: Genetic Association and Possible Pathogenic Roles. Neuromolecular Med 2020; 22:464-473. [PMID: 32894413 DOI: 10.1007/s12017-020-08612-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 08/29/2020] [Indexed: 11/25/2022]
Abstract
Alzheimer's disease (AD) is commonly considered as the most prominent dementing disorder globally and is characterized by the deposition of misfolded amyloid-β (Aβ) peptide and the aggregation of neurofibrillary tangles. Immunological disturbances and neuroinflammation, which result from abnormal immunological reactivations, are believed to be the primary stimulating factors triggering AD-like neuropathy. It has been suggested by multiple previous studies that a bunch of AD key influencing factors might be attributed to genes encoding human leukocyte antigen (HLA), whose variety is an essential part of human adaptive immunity. A wide range of activities involved in immune responses may be determined by HLA genes, including inflammation mediated by the immune response, T-cell transendothelial migration, infection, brain development and plasticity in AD pathogenesis, and so on. The goal of this article is to review the recent epidemiological findings of HLA (mainly HLA class I and II) associated with AD and investigate to what extent the genetic variations of HLA were clinically significant as pathogenic factors for AD. Depending on the degree of contribution of HLA in AD pathogenesis, targeted research towards HLA may propel AD therapeutic strategies into a new era of development.
Collapse
Affiliation(s)
- Zi-Xuan Wang
- Department of Geriatrics, the Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266071, Shandong Province, China.
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, No.308 Ningxia Road, Qingdao, 266071, China.
| | - Qi Wan
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, No.308 Ningxia Road, Qingdao, 266071, China.
- Department of Neurosurgery, Qingdao University, Qingdao, 266071, China.
- Department of Pathophysiology, Qingdao University, Qingdao, 266071, China.
| | - Ang Xing
- Department of Geriatrics, the Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266071, Shandong Province, China
| |
Collapse
|
122
|
Significance of Blood and Cerebrospinal Fluid Biomarkers for Alzheimer's Disease: Sensitivity, Specificity and Potential for Clinical Use. J Pers Med 2020; 10:jpm10030116. [PMID: 32911755 PMCID: PMC7565390 DOI: 10.3390/jpm10030116] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 08/21/2020] [Accepted: 09/01/2020] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia, affecting more than 5 million Americans, with steadily increasing mortality and incredible socio-economic burden. Not only have therapeutic efforts so far failed to reach significant efficacy, but the real pathogenesis of the disease is still obscure. The current theories are based on pathological findings of amyloid plaques and tau neurofibrillary tangles that accumulate in the brain parenchyma of affected patients. These findings have defined, together with the extensive neurodegeneration, the diagnostic criteria of the disease. The ability to detect changes in the levels of amyloid and tau in cerebrospinal fluid (CSF) first, and more recently in blood, has allowed us to use these biomarkers for the specific in-vivo diagnosis of AD in humans. Furthermore, other pathological elements of AD, such as the loss of neurons, inflammation and metabolic derangement, have translated to the definition of other CSF and blood biomarkers, which are not specific of the disease but, when combined with amyloid and tau, correlate with the progression from mild cognitive impairment to AD dementia, or identify patients who will develop AD pathology. In this review, we discuss the role of current and hypothetical biomarkers of Alzheimer's disease, their specificity, and the caveats of current high-sensitivity platforms for their peripheral detection.
Collapse
|
123
|
Preparation and in vitro activity of single chain antibodies against Alzheimer's disease. Immunol Lett 2020; 227:1-7. [PMID: 32781005 DOI: 10.1016/j.imlet.2020.07.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/11/2020] [Accepted: 07/21/2020] [Indexed: 01/16/2023]
Abstract
Disease-modifying passive immunotherapies focused on reducing amyloid-beta (Aβ) deposition are a potential therapeutic strategy for Alzheimer's disease (AD). However, the results of Aβ passive immunotherapy clinical trials were unsatisfactory, largely due to the low efficacy of whole antibodies due to their relatively large molecular weight and low blood-brain barrier transmittance. Furthermore, the constant region fragments of whole antibodies can trigger inflammatory reactions, which raises safety concerns. Single chain fragment variables (scFvs), containing only the variable region of the heavy and light chains of antibodies, show great potential for the treatment of AD. With the aim of generating a safe and effective AD passive immunotherapy, we designed and successfully prepared scFvs targeting Aβ and investigated their activity in vitro. The results showed that both the 10D5-scFv and 12B4-scFv have high affinities for Aβ monomers, oligomers, and fibers. Moreover, scFvs could prevent the formation of Aβ oligomers and fibers, and block their cellular toxicity. In addition, 10D5-scFv and 12B4-scFv could bind to Aβ plaques on the sections of mice brains in the in vitro study, indicating potential for the treatment of AD.
Collapse
|
124
|
Abstract
Individuals with Down syndrome (DS) are at high risk for developing Alzheimer's disease (AD) pathology and this has provided significant insights into our understanding of the genetic basis of AD. The present review summarizes recent clinical, neuropathologic, imaging, and fluid biomarker studies of AD in DS (DSAD), highlighting the striking similarities, as well as some notable differences, between DSAD and the more common late-onset form of AD (LOAD) in the general population, as well as the much rarer, autosomal-dominant form of AD (ADAD). There has been significant progress in our understanding of the natural history of AD biomarkers in DS and their relationship to clinically meaningful changes. Additional work is needed to clearly define the continuum of AD that has been described in the general population, such as the preclinical, prodromal, and dementia stages of AD. Multiple therapeutic approaches, including those targeting not only β-amyloid but also tau and the amyloid precursor protein itself, require consideration. Recent developments in the field are presented within the context of such efforts to conduct clinical trials to treat and potentially prevent AD in DS.
Collapse
Affiliation(s)
- Michael S Rafii
- Alzheimer's Therapeutic Research Institute (ATRI), Keck School of Medicine, University of Southern California, 9860 Mesa Rim Road, San Diego, CA, 92121, USA.
| |
Collapse
|
125
|
Boche D, Nicoll JAR. Invited Review - Understanding cause and effect in Alzheimer's pathophysiology: Implications for clinical trials. Neuropathol Appl Neurobiol 2020; 46:623-640. [PMID: 32643143 DOI: 10.1111/nan.12642] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/23/2020] [Accepted: 07/01/2020] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) pathology is multi-faceted, including extracellular accumulation of amyloid-β (Aβ), accumulation of tau within neurons, glial activation and loss of neurons and synapses. From a neuropathological perspective, usually at a single time-point and often at the end-stage of the disease, it is challenging to understand the cause and effect relationships between these components. There are at least four ways of trying to unravel these relationships. First, genetic studies demonstrate mutations that influence Aβ production, but not tau, can initiate AD; whereas genetic variants influencing AD risk are related to innate immunity and lipid metabolism. Second, studies at early time points show that pathology begins decades before the onset of dementia and indicate different anatomical locations for initiation of Aβ and tau accumulation. Third, cause and effect can be studied in experimental models, but most animal models do not fully replicate AD pathology. However, induced pluripotent stem cells (iPSCs) to study live human neurons has introduced a new perspective. Fourth, clinical trials may alter AD pathology giving insights into cause and effect relationships. Therefore, a sequence of (i) neocortical Aβ accumulation followed by (ii) a microglial inflammatory reaction to Aβ, causing neuritic dystrophy which promotes (iii) spread of tau from the limbic system to the neocortex with (iv) progressive tau accumulation and spread resulting in (v) neurodegeneration, explains the evidence. It is proposed that different therapeutic targets are required for different stages of the disease process: Aβ for primary prevention, microglia for secondary prevention, and tau for established disease.
Collapse
Affiliation(s)
- D Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - J A R Nicoll
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Department of Cellular Pathology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
126
|
Oberman K, Gouweleeuw L, Hoogerhout P, Eisel ULM, van Riet E, Schoemaker RG. Vaccination Prevented Short-Term Memory Loss, but Deteriorated Long-Term Spatial Memory in Alzheimer's Disease Mice, Independent of Amyloid-β Pathology. J Alzheimers Dis Rep 2020; 4:261-280. [PMID: 32904788 PMCID: PMC7458552 DOI: 10.3233/adr-200213] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background: Soluble oligomeric amyloid-β (Aβ), rather than Aβ plaques, seems to be the culprit in Alzheimer’s disease (AD). Accordingly, a new concept vaccine of small cyclic peptide conjugates, selectively targeting oligomeric Aβ, has been developed. Objective: Study the therapeutic potential of this new vaccine in a mouse model for AD. Methods: J20 mice, overexpressing human amyloid precursor protein, were validated for an AD-like phenotype. Then, J20 mice were vaccinated at 2, 3, and 4 months of age and AD phenotype was evaluated at 6, 9, and 12 months of age; or at 9, 10, and 11 months with evaluation at 12 months. Effects on Aβ pathology were studied by plaque load (immunohistochemistry; 6E10) and antibody titers against Aβ (ELISA). AD behavioral phenotype was evaluated by performance in a battery of cognitive tests. Results: J20 mice displayed age-related Aβ plaque development and an AD-like behavioral phenotype. A consistent antibody response to the cyclic peptides was, however, not extended to Aβ, leaving plaque load unaffected. Nevertheless, immunization at young ages prevented working- and short-term spatial memory loss, but deteriorated long-term spatial learning and memory, at 12 months of age. Immunization at later ages did not affect any measured parameter. Conclusion: J20 mice provide a relevant model for AD to study potential anti-Aβ treatment. Early vaccination prevented short-term memory loss at later ages, but deteriorated long-term spatial memory, however without affecting Aβ pathology. Later vaccination had no effects, but optimal timing may require further investigation.
Collapse
Affiliation(s)
- Klaske Oberman
- Department of Neurobiology GELIFES, University Groningen, The Netherlands
| | - Leonie Gouweleeuw
- Department of Neurobiology GELIFES, University Groningen, The Netherlands
| | | | - Ulrich L M Eisel
- Department of Neurobiology GELIFES, University Groningen, The Netherlands
| | | | - Regien G Schoemaker
- Department of Neurobiology GELIFES, University Groningen, The Netherlands.,University Medical Center Groningen, The Netherlands
| |
Collapse
|
127
|
Yang D, Zhu W, Wang Y, Tan F, Ma Z, Gao J, Lin X. Selection of mutant µplasmin for amyloid-β cleavage in vivo. Sci Rep 2020; 10:12117. [PMID: 32694536 PMCID: PMC7374754 DOI: 10.1038/s41598-020-69079-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 07/01/2020] [Indexed: 12/25/2022] Open
Abstract
One of the main culprits of Alzheimer's disease (AD) is the formation of toxic amyloid-β (Aβ) peptide polymers and the aggregation of Aβ to form plaques in the brain. We have developed techniques to purify the catalytic domain of plasmin, micro-plasmin (µPlm), which can be used for an Aβ-clearance based AD therapy. However, in serum, µPlm is irreversibly inhibited by its principal inhibitor α2-antiplasmin (α2-AP). In this study, we engineered and selected mutant forms of µPlm that are both catalytically active and insensitive to α2-AP inhibition. We identified surface residues of μPlm that might interact and bind α2-AP, and used an alanine-scanning mutagenesis method to select residues having higher activity but lower α2-AP inhibition. Then we employed saturation mutagenesis for further optimize both properties. Modeled complex structure of µPlm/α2-AP shows that F587 is a critical contact residue, which can be used as a starting position for further investigation.
Collapse
Affiliation(s)
- Dongying Yang
- Shandong Provincial Key Laboratory of Biophysics, Shandong Key Laboratory in University of Functional Bioresource Utilization, School of Medicine and Nursing, Dezhou University, Daxuexi Road 566#, Dezhou, 253023, Shandong, China
| | - Wei Zhu
- Key Laboratory of State Ethnic Affairs Commission for Biological Technology, College of Life Science, South-Central University for Nationalities, Wuhan, China
| | - Yingjie Wang
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Fangmei Tan
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Zhiping Ma
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Jiali Gao
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
- Department of Chemistry and Supercomputing Institute, University of Minnesota, Minneapolis, MN, 55455, USA.
| | - Xinli Lin
- Key Laboratory of State Ethnic Affairs Commission for Biological Technology, College of Life Science, South-Central University for Nationalities, Wuhan, China.
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| |
Collapse
|
128
|
Iqbal UH, Zeng E, Pasinetti GM. The Use of Antimicrobial and Antiviral Drugs in Alzheimer's Disease. Int J Mol Sci 2020; 21:E4920. [PMID: 32664669 PMCID: PMC7404195 DOI: 10.3390/ijms21144920] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 12/15/2022] Open
Abstract
The aggregation and accumulation of amyloid-β plaques and tau proteins in the brain have been central characteristics in the pathophysiology of Alzheimer's disease (AD), making them the focus of most of the research exploring potential therapeutics for this neurodegenerative disease. With success in interventions aimed at depleting amyloid-β peptides being limited at best, a greater understanding of the physiological role of amyloid-β peptides is needed. The development of amyloid-β plaques has been determined to occur 10-20 years prior to AD symptom manifestation, hence earlier interventions might be necessary to address presymptomatic AD. Furthermore, recent studies have suggested that amyloid-β peptides may play a role in innate immunity as an antimicrobial peptide. These findings, coupled with the evidence of pathogens such as viruses and bacteria in AD brains, suggests that the buildup of amyloid-β plaques could be a response to the presence of viruses and bacteria. This has led to the foundation of the antimicrobial hypothesis for AD. The present review will highlight the current understanding of amyloid-β, and the role of bacteria and viruses in AD, and will also explore the therapeutic potential of antimicrobial and antiviral drugs in Alzheimer's disease.
Collapse
Affiliation(s)
| | | | - Giulio M. Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (U.H.I.); (E.Z.)
| |
Collapse
|
129
|
Caselli RJ, Knopman DS, Bu G. An agnostic reevaluation of the amyloid cascade hypothesis of Alzheimer's disease pathogenesis: The role of APP homeostasis. Alzheimers Dement 2020; 16:1582-1590. [PMID: 32588983 DOI: 10.1002/alz.12124] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 05/03/2020] [Accepted: 05/06/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To reassess the role of amyloid beta (Aβ) and the amyloid precursor protein (APP) system in the pathogenesis of Alzheimer's disease (AD). BACKGROUND APP is a cell adhesion molecule that has been highly conserved over the course of phylogeny that has critical roles in brain development, synaptic plasticity, and the brain's intrinsic immune system. The amyloid cascade hypothesis describes a relatively linear, deterministic sequence of events triggered by a gain of Aβ peptide fragment toxicity that results in neurodegeneration and cognitive loss, yet well designed immunotherapy and beta secretase inhibitor trials that have successfully targeted Aβ have failed to have any consistent effects on the steady decline of cognition. NEW/UPDATED HYPOTHESIS Mutations of the APP and presenilin genes not only alter the ratio of longer to shorter Aβ fragments (resulting in a gain of Aβ toxicity), but also disrupt the normal homeostatic roles of their respective proteins. The evolutionary history, physiological importance, and complexity of the APP and presenilin systems, as well as other critical components including tau and apolipoprotein E (APOE) imply that altered function of such systems could have severe consequences that include but need not be limited to a gain of Aβ toxicity and would more generally result in altered homeostasis of APP-related functions. MAJOR CHALLENGES ADDRESSED BY OUR HYPOTHESIS Challenges that a loss of APP homeostasis addresses better than the more limited gain of Aβ toxicity model include the topographic mismatches between Aβ and tau pathology, the profile and chronology of cognitive and biomarker changes that precede the clinical expression of mild cognitive impairment and dementia, and the disappointments of Aβ targeted therapeutics among others. LINKAGE TO OTHER MAJOR THEORIES The importance of APP, α- and β-secretases, the presenilins and γ-secretase, as well as tau was recognized by the authors of the amyloid cascade hypothesis, and has since led multiple investigators to propose alternative, more balanced hypotheses including reduced homeostasis and frank loss-of-function of key components that include but go beyond the currently envisioned linear model of Aβ toxicity.
Collapse
Affiliation(s)
- Richard J Caselli
- Department of Neurology, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - David S Knopman
- Department of Neurology, Mayo Clinic Rochester, Rochester, Minnesota, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida, USA
| |
Collapse
|
130
|
Khalifeh M, Read MI, Barreto GE, Sahebkar A. Trehalose against Alzheimer's Disease: Insights into a Potential Therapy. Bioessays 2020; 42:e1900195. [PMID: 32519387 DOI: 10.1002/bies.201900195] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 04/13/2020] [Indexed: 12/21/2022]
Abstract
Trehalose is a natural disaccharide with a remarkable ability to stabilize biomolecules. In recent years, trehalose has received growing attention as a neuroprotective molecule and has been tested in experimental models for different neurodegenerative diseases. Although the underlying neuroprotective mechanism of trehalose's action is unclear, one of the most important hypotheses is autophagy induction. The chaperone-like activity of trehalose and the ability to modulate inflammatory responses has also been reported. There is compelling evidence that the dysfunction of autophagy and aggregation of misfolded proteins contribute to the pathogenesis of Alzheimer's disease (AD) and other neurodegenerative disorders. Therefore, given the linking between trehalose and autophagy induction, it appears to be a promising therapy for AD. Herein, the published studies concerning the use of trehalose as a potential therapy for AD are summarized, providing a rationale for applying trehalose to reduce Alzheimer's pathology.
Collapse
Affiliation(s)
- Masoomeh Khalifeh
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Morgayn I Read
- Department of Pharmacology, School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
131
|
Petrushina I, Hovakimyan A, Harahap-Carrillo IS, Davtyan H, Antonyan T, Chailyan G, Kazarian K, Antonenko M, Jullienne A, Hamer MM, Obenaus A, King O, Zagorski K, Blurton-Jones M, Cribbs DH, Lander H, Ghochikyan A, Agadjanyan MG. Characterization and preclinical evaluation of the cGMP grade DNA based vaccine, AV-1959D to enter the first-in-human clinical trials. Neurobiol Dis 2020; 139:104823. [PMID: 32119976 PMCID: PMC8772258 DOI: 10.1016/j.nbd.2020.104823] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 02/03/2020] [Accepted: 02/27/2020] [Indexed: 02/08/2023] Open
Abstract
The DNA vaccine, AV-1959D, targeting N-terminal epitope of Aβ peptide, has been proven immunogenic in mice, rabbits, and non-human primates, while its therapeutic efficacy has been shown in mouse models of Alzheimer's disease (AD). Here we report for the first time on IND-enabling biodistribution and safety/toxicology studies of cGMP-grade AV-1959D vaccine in the Tg2576 mouse model of AD. We also tested acute neuropathology safety profiles of AV-1959D in another AD disease model, Tg-SwDI mice with established vascular and parenchymal Aβ pathology in a pre-clinical translational study. Biodistribution studies two days after the injection demonstrated high copy numbers of AV-1959D plasmid after single immunization of Tg2576 mice at the injection sites but not in the tissues of distant organs. Plasmids persisted at the injection sites of some mice 60 days after vaccination. In Tg2576 mice with established amyloid pathology, we did not observe short- or long-term toxicities after multiple immunizations with three doses of AV-1959D. Assessment of the repeated dose acute safety of AV-1959D in cerebral amyloid angiopathy (CAA) prone Tg-SwDI mice did not reveal any immunotherapy-induced vasogenic edema detected by magnetic resonance imaging (MRI) or increased microhemorrhages. Multiple immunizations of Tg-SwDI mice with AV-1959D did not induce T and B cell infiltration, glial activation, vascular deposition of Aβ, or neuronal degeneration (necrosis and apoptosis) greater than that in the control group determined by immunohistochemistry of brain tissues. Taken together, the safety data from two different mouse models of AD substantiate a favorable safety profile of the cGMP grade AV-1959D vaccine supporting its progression to first-in-human clinical trials.
Collapse
Affiliation(s)
- Irina Petrushina
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Armine Hovakimyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | | | - Hayk Davtyan
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA; Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Tatevik Antonyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Gor Chailyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Konstantin Kazarian
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Maxim Antonenko
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Amandine Jullienne
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Mary M Hamer
- Department of Pediatrics, University of California, Irvine, CA, USA
| | - Andre Obenaus
- Department of Pediatrics, University of California, Irvine, CA, USA; Preclinical and Translational Imaging Center, University of California, Irvine, CA, USA
| | - Olga King
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Karen Zagorski
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Mathew Blurton-Jones
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA; Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - David H Cribbs
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Harry Lander
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA
| | - Anahit Ghochikyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA.
| | - Michael G Agadjanyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA, USA.
| |
Collapse
|
132
|
Ordóñez-Gutiérrez L, Wandosell F. Nanoliposomes as a Therapeutic Tool for Alzheimer's Disease. Front Synaptic Neurosci 2020; 12:20. [PMID: 32523525 PMCID: PMC7261886 DOI: 10.3389/fnsyn.2020.00020] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/24/2020] [Indexed: 12/31/2022] Open
Abstract
The accumulation of extracellular amyloid-beta (Aβ), denoted as senile plaques, and intracellular neurofibrillary tangles (formed by hyperphosphorylated Tau protein) in the brain are two major neuropathological hallmarks of Alzheimer's disease (AD). The current and most accepted hypothesis proposes that the oligomerization of Aβ peptides triggers the polymerization and accumulation of amyloid, which leads to the senile plaques. Several strategies have been reported to target Aβ oligomerization/polymerization. Since it is thought that Aβ levels in the brain and peripheral blood maintain equilibrium, it has been hypothesized that enhancing peripheral clearance (by shifting this equilibrium towards the blood) might reduce Aβ levels in the brain, known as the sink effect. This process has been reported to be effective, showing a reduction in Aβ burden in the brain as a consequence of the peripheral reduction of Aβ levels. Nanoparticles (NPs) may have difficulty crossing the blood-brain barrier (BBB), initially due to their size. It is not clear whether particles in the range of 50-100 nm should be able to cross the BBB without being specifically modified for it. Despite the size limitation of crossing the BBB, several NP derivatives may be proposed as therapeutic tools. The purpose of this review is to summarize some therapeutic approaches based on nanoliposomes using two complementary examples: First, unilamellar nanoliposomes containing Aβ generic ligands, such as sphingolipids, gangliosides or curcumin, or some sphingolipid bound to the binding domain of ApoE; and second, nanoliposomes containing monoclonal antibodies against Aβ. Following similar rationale NPs of poly(lactide-co-glycolide)-poly (ethylene glycol) conjugated with curcumin-derivate (PLGA-PEG-B6/Cur) were reported to improve the spatial learning and memory capability of APP/PS1 mice, compared with native curcumin treatment. Also, some new nanostructures such as exosomes have been proposed as a putative therapeutic and prevention strategies of AD. Although the unquestionable interest of this issue is beyond the scope of this review article. The potential mechanisms and significance of nanoliposome therapies for AD, which are still are in clinical trials, will be discussed.
Collapse
Affiliation(s)
- Lara Ordóñez-Gutiérrez
- Department of Molecular Neurobiology, Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Universidad Autónoma Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Francisco Wandosell
- Department of Molecular Neurobiology, Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Universidad Autónoma Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
133
|
Nicoll JAR, Buckland GR, Harrison CH, Page A, Harris S, Love S, Neal JW, Holmes C, Boche D. Persistent neuropathological effects 14 years following amyloid-β immunization in Alzheimer's disease. Brain 2020; 142:2113-2126. [PMID: 31157360 PMCID: PMC6598630 DOI: 10.1093/brain/awz142] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 03/20/2019] [Accepted: 04/04/2019] [Indexed: 11/29/2022] Open
Abstract
We performed a 15-year post-mortem neuropathological follow-up of patients in the first trial of amyloid-β immunotherapy for Alzheimer’s disease. Twenty-two participants of a clinical trial of active amyloid-β42 immunization (AN1792, Elan Pharmaceuticals) or placebo were studied. Comprehensive post-mortem neuropathological assessments were performed from 4 months to 15 years after the trial. We analysed the relationships between the topographical distribution of amyloid-β removal from the cerebral cortex and tau pathology, cerebrovascular territories, plasma anti-AN1792 antibody titres and late cognitive status. Seventeen of 22 (77%) participants had Alzheimer’s neuropathological change, whereas 5 of 22 (23%) had alternative causes for dementia (progressive supranuclear palsy = 1, Lewy body disease = 1, vascular brain injury = 1, and frontotemporal lobar degeneration = 2). Nineteen of the 22 participants had received the active agent, three the placebo. Fourteen of 16 (88%) patients with Alzheimer’s disease receiving the active agent had evidence of plaque removal (very extensive removal = 5, intermediate = 4, very limited = 5, no removal = 2). Of particular note, two Alzheimer’s patients who died 14 years after immunization had only very sparse or no detectable plaques in all regions examined. There was a significant inverse correlation between post-vaccination peripheral blood anti-AN1792 antibody titres and post-mortem plaque scores (ρ = − 0.664, P = 0.005). Cortical foci cleared of plaques contained less tau than did cortex with remaining plaques, but the overall distribution of tangles was extensive (Braak V/VI). In conclusion, patients with Alzheimer’s disease actively immunized against amyloid-β can remain virtually plaque-free for 14 years. The extent of plaque removal is related to the immune response. This long duration of efficacy is important in support of active immunization protocols as therapy for, or potentially prevention of, neurodegeneration-associated protein accumulations. Inclusion of patients without Alzheimer’s disease in Alzheimer’s therapy trials is a problem for assessing the efficacy of treatment. Despite modification of Alzheimer’s pathology, most patients had progressed to severe dementia, notably including the five with very extensive plaque removal, possibly due to continued tau propagation. Neuropathology follow-up of patients in therapeutic trials provides valuable information on the causes of dementia and effects of treatment.
Collapse
Affiliation(s)
- James A R Nicoll
- Clinical Neurosciences, Clinical and Experimental Sciences, University of Southampton, Southampton, UK.,Department of Cellular Pathology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - George R Buckland
- Clinical Neurosciences, Clinical and Experimental Sciences, University of Southampton, Southampton, UK
| | - Charlotte H Harrison
- Clinical Neurosciences, Clinical and Experimental Sciences, University of Southampton, Southampton, UK
| | - Anton Page
- Biomedical Imaging Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Scott Harris
- Public Health Sciences and Medical Statistics, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Seth Love
- Department of Neuropathology, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - James W Neal
- Department of Cellular Pathology, University Hospital of Wales, Cardiff University, Cardiff, UK
| | - Clive Holmes
- Clinical Neurosciences, Clinical and Experimental Sciences, University of Southampton, Southampton, UK.,Memory Assessment and Research Centre, Moorgreen Hospital, Southern Health Foundation Trust, Southampton, UK
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
134
|
Mantile F, Capasso A, De Berardinis P, Prisco A. Analysis of the Consolidation Phase of Immunological Memory within the IgG Response to a B Cell Epitope Displayed on a Filamentous Bacteriophage. Microorganisms 2020; 8:564. [PMID: 32295280 PMCID: PMC7232419 DOI: 10.3390/microorganisms8040564] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/07/2020] [Accepted: 04/10/2020] [Indexed: 02/06/2023] Open
Abstract
Immunological memory can be defined as the ability to mount a response of greater magnitude and with faster kinetics upon re-encounter of the same antigen. We have previously reported that a booster dose of a protein antigen given 15 days after the first dose interferes with the development of memory, i.e., with the ability to mount an epitope-specific IgG response of greater magnitude upon re-encounter of the same antigen. We named the time-window during which memory is vulnerable to disruption a "consolidation phase in immunological memory", by analogy with the memory consolidation processes that occur in the nervous system to stabilize memory traces. In this study, we set out to establish if a similar memory consolidation phase occurs in the IgG response to a B cell epitope displayed on a filamentous bacteriophage. To this end, we have analyzed the time-course of anti-β-amyloid IgG titers in mice immunized with prototype Alzheimer's Disease vaccine fdAD(2-6), which consists of a fd phage that displays the B epitope AEFRH of β -amyloid at the N-terminus of the Major Capsid Protein. A booster dose of phage fdAD(2-6) given 15 days after priming significantly reduced the ratio between the magnitude of the secondary and primary IgG response to β-amyloid. This analysis confirms, in a phage vaccine, a consolidation phase in immunological memory, occurring two weeks after priming.
Collapse
Affiliation(s)
- Francesca Mantile
- Institute of Genetics and Biophysics, CNR, 80131 Naples, Italy; (F.M.); (A.C.)
| | - Angelo Capasso
- Institute of Genetics and Biophysics, CNR, 80131 Naples, Italy; (F.M.); (A.C.)
| | | | - Antonella Prisco
- Institute of Genetics and Biophysics, CNR, 80131 Naples, Italy; (F.M.); (A.C.)
| |
Collapse
|
135
|
Muscat S, Pallante L, Stojceski F, Danani A, Grasso G, Deriu MA. The Impact of Natural Compounds on S-Shaped Aβ42 Fibril: From Molecular Docking to Biophysical Characterization. Int J Mol Sci 2020; 21:ijms21062017. [PMID: 32188076 PMCID: PMC7139307 DOI: 10.3390/ijms21062017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/06/2020] [Accepted: 03/13/2020] [Indexed: 12/18/2022] Open
Abstract
The pursuit for effective strategies inhibiting the amyloidogenic process in neurodegenerative disorders, such as Alzheimer’s disease (AD), remains one of the main unsolved issues, and only a few drugs have demonstrated to delay the degeneration of the cognitive system. Moreover, most therapies induce severe side effects and are not effective at all stages of the illness. The need to find novel and reliable drugs appears therefore of primary importance. In this context, natural compounds have shown interesting beneficial effects on the onset and progression of neurodegenerative diseases, exhibiting a great inhibitory activity on the formation of amyloid aggregates and proving to be effective in many preclinical and clinical studies. However, their inhibitory mechanism is still unclear. In this work, ensemble docking and molecular dynamics simulations on S-shaped Aβ42 fibrils have been carried out to evaluate the influence of several natural compounds on amyloid conformational behaviour. A deep understanding of the interaction mechanisms between natural compounds and Aβ aggregates may play a key role to pave the way for design, discovery and optimization strategies toward an efficient destabilization of toxic amyloid assemblies.
Collapse
Affiliation(s)
- Stefano Muscat
- Dalle Molle Institute for Artificial Intelligence (IDSIA), University of Italian Switzerland (USI), University of Applied Science and Art of Southern Switzerland (SUPSI), CH-6928 Manno, Switzerland
| | - Lorenzo Pallante
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, IT-10128 Torino, Italy
| | - Filip Stojceski
- Dalle Molle Institute for Artificial Intelligence (IDSIA), University of Italian Switzerland (USI), University of Applied Science and Art of Southern Switzerland (SUPSI), CH-6928 Manno, Switzerland
| | - Andrea Danani
- Dalle Molle Institute for Artificial Intelligence (IDSIA), University of Italian Switzerland (USI), University of Applied Science and Art of Southern Switzerland (SUPSI), CH-6928 Manno, Switzerland
| | - Gianvito Grasso
- Dalle Molle Institute for Artificial Intelligence (IDSIA), University of Italian Switzerland (USI), University of Applied Science and Art of Southern Switzerland (SUPSI), CH-6928 Manno, Switzerland
| | - Marco Agostino Deriu
- PolitoBIOMed Lab, Department of Mechanical and Aerospace Engineering, Politecnico di Torino, IT-10128 Torino, Italy
- Correspondence:
| |
Collapse
|
136
|
Tauopathy Analysis in P301S Mouse Model of Alzheimer Disease Immunized With DNA and MVA Poxvirus-Based Vaccines Expressing Human Full-Length 4R2N or 3RC Tau Proteins. Vaccines (Basel) 2020; 8:vaccines8010127. [PMID: 32183198 PMCID: PMC7157204 DOI: 10.3390/vaccines8010127] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/10/2020] [Accepted: 03/12/2020] [Indexed: 01/04/2023] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by a progressive memory loss and cognitive decline that has been associated with an accumulation in the brain of intracellular neurofibrillary tangles (NFTs) formed by hyperphosphorylated tau protein, and extracellular senile plaques formed by β-amyloid peptides. Currently, there is no cure for AD and after the failure of anti β-amyloid therapies, active and passive tau immunotherapeutic approaches have been developed in order to prevent, reduce or ideally reverse the disease. Vaccination is one of the most effective approaches to prevent diseases and poxviruses, particularly modified vaccinia virus Ankara (MVA), are one of the most promising viral vectors used as vaccines against several human diseases. Thus, we present here the generation and characterization of the first MVA vectors expressing human tau genes; the full-length 4R2N tau protein or a 3RC tau fragment containing 3 tubulin-binding motifs and the C-terminal region (termed MVA-Tau4R2N and MVA-Tau3RC, respectively). Both MVA-Tau recombinant viruses efficiently expressed the human tau 4R2N or 3RC proteins in cultured cells, being detected in the cytoplasm of infected cells and co-localized with tubulin. These MVA-Tau vaccines impacted the innate immune responses with a differential recruitment of innate immune cells to the peritoneal cavity of infected mice. However, no tau-specific T cell or humoral immune responses were detected in vaccinated mice. Immunization of transgenic P301S mice, a mouse model for tauopathies, with a DNA-Tau prime/MVA-Tau boost approach showed no significant differences in the hyperphosphorylation of tau, motor capacity and survival rate, when compared to non-vaccinated mice. These findings showed that a well-established and potent protocol of T and B cell activation based on DNA/MVA prime/boost regimens using DNA and MVA vectors expressing tau full-length 4R2N or 3RC proteins is not sufficient to trigger tau-specific T and B cell immune responses and to induce a protective effect against tauopathy in this P301S murine model. In the pursuit of AD vaccines, our results highlight the need for novel optimized tau immunogens and additional modes of presentation of tau protein to the immune system.
Collapse
|
137
|
Zhou Q, Mareljic N, Michaelsen M, Parhizkar S, Heindl S, Nuscher B, Farny D, Czuppa M, Schludi C, Graf A, Krebs S, Blum H, Feederle R, Roth S, Haass C, Arzberger T, Liesz A, Edbauer D. Active poly-GA vaccination prevents microglia activation and motor deficits in a C9orf72 mouse model. EMBO Mol Med 2020; 12:e10919. [PMID: 31858749 PMCID: PMC7005532 DOI: 10.15252/emmm.201910919] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 11/29/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022] Open
Abstract
The C9orf72 repeat expansion is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and/or frontotemporal dementia (FTD). Non-canonical translation of the expanded repeat results in abundant poly-GA inclusion pathology throughout the CNS. (GA)149 -CFP expression in mice triggers motor deficits and neuroinflammation. Since poly-GA is transmitted between cells, we investigated the therapeutic potential of anti-GA antibodies by vaccinating (GA)149 -CFP mice. To overcome poor immunogenicity, we compared the antibody response of multivalent ovalbumin-(GA)10 conjugates and pre-aggregated carrier-free (GA)15 . Only ovalbumin-(GA)10 immunization induced a strong anti-GA response. The resulting antisera detected poly-GA aggregates in cell culture and patient tissue. Ovalbumin-(GA)10 immunization largely rescued the motor function in (GA)149 -CFP transgenic mice and reduced poly-GA inclusions. Transcriptome analysis showed less neuroinflammation in ovalbumin-(GA)10 -immunized poly-GA mice, which was corroborated by semiquantitative and morphological analysis of microglia/macrophages. Moreover, cytoplasmic TDP-43 mislocalization and levels of the neurofilament light chain in the CSF were reduced, suggesting neuroaxonal damage is reduced. Our data suggest that immunotherapy may be a viable primary prevention strategy for ALS/FTD in C9orf72 mutation carriers.
Collapse
Affiliation(s)
- Qihui Zhou
- German Center for Neurodegenerative Diseases (DZNE), MunichMunichGermany
- Munich Cluster of Systems Neurology (SyNergy)MunichGermany
| | - Nikola Mareljic
- German Center for Neurodegenerative Diseases (DZNE), MunichMunichGermany
| | - Meike Michaelsen
- German Center for Neurodegenerative Diseases (DZNE), MunichMunichGermany
| | - Samira Parhizkar
- Chair of Metabolic BiochemistryBiomedical Center (BMC)Faculty of MedicineLudwig‐Maximilians‐Universität MunichMunichGermany
| | - Steffanie Heindl
- Institute for Stroke and Dementia ResearchLudwig‐Maximilians‐University MunichMunichGermany
| | - Brigitte Nuscher
- Chair of Metabolic BiochemistryBiomedical Center (BMC)Faculty of MedicineLudwig‐Maximilians‐Universität MunichMunichGermany
| | - Daniel Farny
- German Center for Neurodegenerative Diseases (DZNE), MunichMunichGermany
| | - Mareike Czuppa
- German Center for Neurodegenerative Diseases (DZNE), MunichMunichGermany
| | - Carina Schludi
- German Center for Neurodegenerative Diseases (DZNE), MunichMunichGermany
| | - Alexander Graf
- Laboratory for Functional Genome AnalysisGene CenterLudwig Maximilian University of MunichMunichGermany
| | - Stefan Krebs
- Laboratory for Functional Genome AnalysisGene CenterLudwig Maximilian University of MunichMunichGermany
| | - Helmut Blum
- Laboratory for Functional Genome AnalysisGene CenterLudwig Maximilian University of MunichMunichGermany
| | - Regina Feederle
- German Center for Neurodegenerative Diseases (DZNE), MunichMunichGermany
- Munich Cluster of Systems Neurology (SyNergy)MunichGermany
- Monoclonal Antibody Core Facility and Research GroupInstitute for Diabetes and ObesityHelmholtz Zentrum MünchenGerman Research Center for Environmental Health (GmbH)MunichGermany
| | - Stefan Roth
- Munich Cluster of Systems Neurology (SyNergy)MunichGermany
- Institute for Stroke and Dementia ResearchLudwig‐Maximilians‐University MunichMunichGermany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), MunichMunichGermany
- Munich Cluster of Systems Neurology (SyNergy)MunichGermany
- Chair of Metabolic BiochemistryBiomedical Center (BMC)Faculty of MedicineLudwig‐Maximilians‐Universität MunichMunichGermany
| | - Thomas Arzberger
- German Center for Neurodegenerative Diseases (DZNE), MunichMunichGermany
- Munich Cluster of Systems Neurology (SyNergy)MunichGermany
- Center for Neuropathology and Prion ResearchLudwig‐Maximilians‐University MunichMunichGermany
- Department of Psychiatry and PsychotherapyUniversity HospitalLudwig‐Maximilians‐University MunichMunichGermany
| | - Arthur Liesz
- Munich Cluster of Systems Neurology (SyNergy)MunichGermany
- Institute for Stroke and Dementia ResearchLudwig‐Maximilians‐University MunichMunichGermany
| | - Dieter Edbauer
- German Center for Neurodegenerative Diseases (DZNE), MunichMunichGermany
- Munich Cluster of Systems Neurology (SyNergy)MunichGermany
- Ludwig‐Maximilians‐University MunichMunichGermany
| |
Collapse
|
138
|
Aliseychik M, Patrikeev A, Gusev F, Grigorenko A, Andreeva T, Biragyn A, Rogaev E. Dissection of the Human T-Cell Receptor γ Gene Repertoire in the Brain and Peripheral Blood Identifies Age- and Alzheimer's Disease-Associated Clonotype Profiles. Front Immunol 2020; 11:12. [PMID: 32117220 PMCID: PMC7025544 DOI: 10.3389/fimmu.2020.00012] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 01/06/2020] [Indexed: 01/08/2023] Open
Abstract
The immune system contributes to neurodegenerative pathologies. However, the roles of γδ T cells in Alzheimer's disease (AD) are poorly understood. Here, we evaluated somatic variability of T-cell receptor γ genes (TRGs) in patients with AD. We performed deep sequencing of the CDR3 region of TRGs in patients with AD and control patients without dementia. TRG clones were clearly detectable in peripheral blood (PB) and non-neuronal cell populations in human brains. TRG repertoire diversity was reduced during aging. Compared with the PB, the brain showed reduced TRGV9 clonotypes but was enriched in TRGV2/4/8 clonotypes. AD-associated TRG profiles were found in both the PB and brain. Moreover, some groups of clonotypes were more specific for the brain or blood in patients with AD compared to those in controls. Our pilot deep analysis of T-cell receptor diversities in AD revealed putative brain and AD-associated immunogenic markers.
Collapse
Affiliation(s)
- Maria Aliseychik
- Department of Psychiatry, University of Massachusetts Medical School, Worcester, MA, United States.,Department of Human Genetics and Genomics, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia.,Center for Genetics and Genetic Technologies, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Anton Patrikeev
- Department of Human Genetics and Genomics, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Fedor Gusev
- Department of Human Genetics and Genomics, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Anastasia Grigorenko
- Department of Psychiatry, University of Massachusetts Medical School, Worcester, MA, United States.,Department of Human Genetics and Genomics, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Tatiana Andreeva
- Department of Human Genetics and Genomics, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia.,Center for Genetics and Genetic Technologies, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Arya Biragyn
- Immunoregulation Section, National Institute on Aging, Baltimore, MD, United States
| | - Evgeny Rogaev
- Department of Psychiatry, University of Massachusetts Medical School, Worcester, MA, United States.,Department of Human Genetics and Genomics, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia.,Center for Genetics and Genetic Technologies, Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
139
|
Ní Chasaide C, Lynch MA. The role of the immune system in driving neuroinflammation. Brain Neurosci Adv 2020; 4:2398212819901082. [PMID: 32219178 PMCID: PMC7085916 DOI: 10.1177/2398212819901082] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation is now recognised as an important contributory factor in the progression of Alzheimer’s disease and probably also in the early stages of the disease. It is likely that this derives largely from aberrant activation of microglia, the resident mononuclear phagocytes of the brain. These cells are responsible for physiological immune surveillance and clearance of pathogens in the central nervous system, but evidence indicates that in Alzheimer’s disease, microglial function is compromised, and this contributes to the pathology. It is unclear what factors cause the inappropriate activation of the microglia in Alzheimer’s disease, but one contributor may be infiltrating peripheral immune cells and these include macrophages and T cells. It has been suggested that both cell types modulate the phenotype of microglia, highlighting the importance of crosstalk between the innate and adaptive immune system in Alzheimer’s disease. This review outlines our current knowledge of how cells of the peripheral immune system, specifically macrophages and T cells, may modulate microglial phenotype in the context of Alzheimer’s disease and considers the impact on their function, especially phagocytic capacity.
Collapse
Affiliation(s)
| | - Marina A Lynch
- Marina A Lynch, Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
140
|
Elucidating the Effect of Static Electric Field on Amyloid Beta 1-42 Supramolecular Assembly. J Mol Graph Model 2020; 96:107535. [PMID: 31978828 DOI: 10.1016/j.jmgm.2020.107535] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 12/06/2019] [Accepted: 01/09/2020] [Indexed: 02/07/2023]
Abstract
Amyloid-β (Aβ) aggregation is recognized to be a key toxic factor in the pathogenesis of Alzheimer disease, which is the most common progressive neurodegenerative disorder. In vitro experiments have elucidated that Aβ aggregation depends on several factors, such as pH, temperature and peptide concentration. Despite the research effort in this field, the fundamental mechanism responsible for the disease progression is still unclear. Recent research has proposed the application of electric fields as a non-invasive therapeutic option leading to the disruption of amyloid fibrils. In this regard, a molecular level understanding of the interactions governing the destabilization mechanism represents an important research advancement. Understanding the electric field effects on proteins, provides a more in-depth comprehension of the relationship between protein conformation and electrostatic dipole moment. The present study focuses on investigating the effect of static Electric Field (EF) on the conformational dynamics of Aβ fibrils by all-atom Molecular Dynamics (MD) simulations. The outcome of this work provides novel insight into this research field, demonstrating how the Aβ assembly may be destabilized by the applied EF.
Collapse
|
141
|
Huang YM, Hong XZ, Shen J, Geng LJ, Pan YH, Ling W, Zhao HL. Amyloids in Site-Specific Autoimmune Reactions and Inflammatory Responses. Front Immunol 2020; 10:2980. [PMID: 31993048 PMCID: PMC6964640 DOI: 10.3389/fimmu.2019.02980] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 12/04/2019] [Indexed: 12/15/2022] Open
Abstract
Amyloid deposition is a histological hallmark of common human disorders including Alzheimer's disease (AD) and type 2 diabetes. Although some reports highlight that amyloid fibrils might activate the innate immunity system via pattern recognition receptors, here, we provide multiple lines of evidence for the protection by site-specific amyloid protein analogs and fibrils against autoimmune attacks: (1) strategies targeting clearance of the AD-related brain amyloid plaque induce high risk of deadly autoimmune destructions in subjects with cognitive dysfunction; (2) administration of amyloidogenic peptides with either full length or core hexapeptide structure consistently ameliorates signs of experimental autoimmune encephalomyelitis; (3) experimental autoimmune encephalomyelitis is exacerbated following genetic deletion of amyloid precursor proteins; (4) absence of islet amyloid coexists with T-cell-mediated insulitis in autoimmune diabetes and autoimmune polyendocrine syndrome; (5) use of islet amyloid polypeptide agonists rather than antagonists improves diabetes care; and (6) common suppressive signaling pathways by regulatory T cells are activated in both local and systemic amyloidosis. These findings indicate dual modulation activity mediated by amyloid protein monomers, oligomers, and fibrils to maintain immune homeostasis. The protection from autoimmune destruction by amyloid proteins offers a novel therapeutic approach to regenerative medicine for common degenerative diseases.
Collapse
Affiliation(s)
- Yan-Mei Huang
- Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Guilin, China.,Center for Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, China
| | - Xue-Zhi Hong
- Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Guilin, China.,Department of Rheumatology and Immunology, The First Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Jian Shen
- Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Guilin, China.,Department of Pathology, The First Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Li-Jun Geng
- Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Guilin, China.,Center for Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, China
| | - Yan-Hong Pan
- Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Guilin, China.,Center for Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, China
| | - Wei Ling
- Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Guilin, China.,Department of Endocrinology, Xiangya Medical School, Central South University, Changsha, China
| | - Hai-Lu Zhao
- Department of Immunology, Guangxi Area of Excellence, Guilin Medical University, Guilin, China.,Center for Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, China.,Institute of Basic Medical Sciences, Faculty of Basic Medicine, Guilin Medical University, Guilin, China
| |
Collapse
|
142
|
Wang JC, Zhu K, Zhang HY, Wang GQ, Liu HY, Cao YP. Early active immunization with Aβ 3-10-KLH vaccine reduces tau phosphorylation in the hippocampus and protects cognition of mice. Neural Regen Res 2020; 15:519-527. [PMID: 31571664 PMCID: PMC6921334 DOI: 10.4103/1673-5374.266061] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Active and passive anti-Aβ immunotherapies have successfully been used for the prevention and treatment of Alzheimer’s disease animal models. However, clinical use of these immunotherapies is not effective, because the vaccination is administered too late. At 1 month of age, 100 μL of Aβ3–10-KLH peptide (vaccine, 2 μg/μL) was subcutaneously injected into the neck of an amyloid precursor protein/presenilin-1/tau transgenic (3×Tg-AD) mouse model. Aβ3–10-KLH peptide was re-injected at 1.5, 2.5, 3.5, 4.5, 5.5, and 6.5 months of age. Serum levels of Aβ antibody were detected by enzyme-linked immunosorbent assay, while spatial learning and memory ability were evaluated by Morris water maze. Immunohistochemistry was used to detect total tau with HT7 and phosphorylated tau with AT8 (phosphorylation sites Ser202 and Thr205) and AT180 (phosphorylation site Thr231) antibodies in the hippocampus. In addition, western blot analysis was used to quantify AT8 and AT180 expression in the hippocampus. The results showed that after vaccine injection, mice produced high levels of Aβ antibody, cognitive function was significantly improved, and total tau and phosphorylated tau levels were significantly reduced. These findings suggest that early active immunization with Aβ3–10-KLH vaccine can greatly reduce tau phosphorylation, thereby mitigating the cognitive decline of 3×Tg-AD mice. This study was approved by the Animal Ethics Committee of China Medical University, China (approval No. 103-316) on April 2, 2016.
Collapse
Affiliation(s)
- Jin-Chun Wang
- Department of Neurology, the Fifth People's Hospital of Shenyang, Shenyang, Liaoning Province, China
| | - Kun Zhu
- Department of Neurology, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Hui-Yi Zhang
- Department of Neurology, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Guo-Qing Wang
- Department of Neurology, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Hui-Ying Liu
- Department of Neurology, the Fifth People's Hospital of Shenyang, Shenyang, Liaoning Province, China
| | - Yun-Peng Cao
- Department of Neurology, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
143
|
Takeda M, Tagami S. A History of Antidementic Drug Development in Japan. TAIWANESE JOURNAL OF PSYCHIATRY 2020. [DOI: 10.4103/tpsy.tpsy_33_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
144
|
Pampuscenko K, Morkuniene R, Sneideris T, Smirnovas V, Budvytyte R, Valincius G, Brown GC, Borutaite V. Extracellular tau induces microglial phagocytosis of living neurons in cell cultures. J Neurochem 2019; 154:316-329. [PMID: 31834946 DOI: 10.1111/jnc.14940] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 02/06/2023]
Abstract
Tau is a microtubule-associated protein, found at high levels in neurons, and its aggregation is associated with neurodegeneration. Recently, it was found that tau can be actively secreted from neurons, but the effects of extracellular tau on neuronal viability are unclear. In this study, we investigated whether extracellular tau2N4R can cause neurotoxicity in primary cultures of rat brain neurons and glial cells. Cell cultures were examined for neuronal loss, death, and phosphatidylserine exposure, as well as for microglial phagocytosis by fluorescence microscopy. Aggregation of tau2N4R was assessed by atomic force microscopy. We found that extracellular addition of tau induced a gradual loss of neurons over 1-2 days, without neuronal necrosis or apoptosis, but accompanied by proliferation of microglia in the neuronal-glial co-cultures. Tau addition caused exposure of the 'eat-me' signal phosphatidylserine on the surface of living neurons, and this was prevented by elimination of the microglia or by inhibition of neutral sphingomyelinase. Tau also increased the phagocytic activity of pure microglia, and this was blocked by inhibitors of neutral sphingomyelinase or protein kinase C. The neuronal loss induced by tau was prevented by inhibitors of neutral sphingomyelinase, protein kinase C or the phagocytic receptor MerTK, or by eliminating microglia from the cultures. The data suggest that extracellular tau induces primary phagocytosis of stressed neurons by activated microglia, and identifies multiple ways in which the neuronal loss induced by tau can be prevented.
Collapse
Affiliation(s)
- Katryna Pampuscenko
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Ramune Morkuniene
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Tomas Sneideris
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Vytautas Smirnovas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Rima Budvytyte
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Gintaras Valincius
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Vilmante Borutaite
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
145
|
Ishima Y, Mimono A, Tuan Giam Chuang V, Fukuda T, Kusumoto K, Okuhira K, Suwa Y, Watanabe H, Ishida T, Morioka H, Maruyama T, Otagiri M. Albumin domain mutants with enhanced Aβ binding capacity identified by phage display analysis for application in various peripheral Aβ elimination approaches of Alzheimer's disease treatment. IUBMB Life 2019; 72:641-651. [PMID: 31794135 DOI: 10.1002/iub.2203] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 11/03/2019] [Indexed: 11/11/2022]
Abstract
Deposition of amyloid protein, particularly Aβ1-42 , is a major contributor to the onset of Alzheimer's disease (AD). However, almost no deposition of Aβ in the peripheral tissues could be found. Human serum albumin (HSA), the most abundant protein in the blood, has been reported to inhibit amyloid formation through binding Aβ, which is believed to play an important role in the peripheral clearance of Aβ. We identified the Aβ binding site on HSA and developed HSA mutants with high binding capacities for Aβ using a phage display method. HSA fragment 187-385 (Domain II) was found to exhibit the highest binding capacity for Aβ compared with the other two HSA fragments. To elucidate the sequence that forms the binding site for Aβ on Domain II, a random screening of Domain II display phage biopanning was constructed. A number of mutants with higher Aβ binding capacities than the wild type were identified. These mutants exhibited stronger scavenging abilities than the wild type, as revealed via in vitro equilibrium dialysis of Aβ experiments. These findings provide useful basic data for developing a safer alternative therapy than Aβ vaccines and for application in plasma exchange as well as extracorporeal dialysis.
Collapse
Affiliation(s)
- Yu Ishima
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan.,School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Subang Jaya, Selangor, Malaysia
| | - Ai Mimono
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Oe-honmachi, Kumamoto, Japan
| | - Victor Tuan Giam Chuang
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Subang Jaya, Selangor, Malaysia
| | - Tetsuya Fukuda
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Oe-honmachi, Kumamoto, Japan
| | - Kohshi Kusumoto
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Keiichiro Okuhira
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Yoshiaki Suwa
- Department of Analytical and Biophysical Chemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroshi Watanabe
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Oe-honmachi, Kumamoto, Japan
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Hiroshi Morioka
- Department of Analytical and Biophysical Chemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Oe-honmachi, Kumamoto, Japan
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Sojo University, Ikeda, Kumamoto, Japan.,DDS Research Institute, Sojo University, Ikeda, Kumamoto, Japan
| |
Collapse
|
146
|
Antibody-based therapies for Huntington’s disease: current status and future directions. Neurobiol Dis 2019; 132:104569. [DOI: 10.1016/j.nbd.2019.104569] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 08/01/2019] [Accepted: 08/02/2019] [Indexed: 12/12/2022] Open
|
147
|
Joly-Amado A, Davtyan H, Serraneau K, Jules P, Zitnyar A, Pressman E, Zagorski K, Antonyan T, Hovakimyan A, Paek HJ, Gordon MN, Cribbs DH, Petrovsky N, Agadjanyan MG, Ghochikyan A, Morgan D. Active immunization with tau epitope in a mouse model of tauopathy induced strong antibody response together with improvement in short memory and pSer396-tau pathology. Neurobiol Dis 2019; 134:104636. [PMID: 31629891 DOI: 10.1016/j.nbd.2019.104636] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/30/2019] [Accepted: 10/07/2019] [Indexed: 12/16/2022] Open
Abstract
Abnormal tau hyperphosphorylation and its aggregation into neurofibrillary tangles are a hallmark of tauopathies, neurodegenerative disorders that include Alzheimer's disease (AD). Active and passive Tau-immunotherapy has been proposed as a therapeutic approach to AD with mixed results. One of the limitations of active immunotherapy may be associated with the mediocre immunogenicity of vaccines that are not inducing therapeutically potent titers of antibodies. The aim of this study was to test the efficacy of an anti-tau vaccine, AV-1980R/A composed of N terminal peptide of this molecule fused with an immunogenic MultiTEP platform and formulated in a strong adjuvant, AdvaxCpG in a Tg4510 mouse model of tauopathy. Experimental mice were immunized with AV-1980R/A and a control group of mice were injected with adjuvant only. Nontransgenic and tetracycline transactivator (tTA) transgenic littermates were included as baseline controls to contrast with the tau phenotype. Active immunization with AV-1980R/A induced very strong anti-tau humoral immune responses in both nontransgenic and transgenic mice with evidence of IgG in brains of AV-1980R/A vaccinated mice. These experimental animals displayed an improvement in short-term memory during a novel object recognition test. However, impairments in other behavioral tasks were not prevented by AV-1980R/A vaccinations. At the same time, high titers of anti-tau antibodies reduced hyperphosphorylated pSer396 tau but did not lower the level of other phosphorylated tau species in the brains of AV-1980R/A vaccinated mice. These data indicate that active immunotherapy with an N-terminal Tau epitope was only partially effective in improving cognition and reducing pathology in the stringent Tg4510 mouse model of tauopathy.
Collapse
Affiliation(s)
- A Joly-Amado
- USF Health Byrd Alzheimer's Institute, Tampa, FL 33613, USA.
| | - H Davtyan
- The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA
| | - K Serraneau
- USF Health Byrd Alzheimer's Institute, Tampa, FL 33613, USA
| | - P Jules
- USF Health Byrd Alzheimer's Institute, Tampa, FL 33613, USA
| | - A Zitnyar
- USF Health Byrd Alzheimer's Institute, Tampa, FL 33613, USA
| | - E Pressman
- USF Health Byrd Alzheimer's Institute, Tampa, FL 33613, USA
| | - K Zagorski
- The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA
| | - T Antonyan
- The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA
| | - A Hovakimyan
- The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA
| | - H J Paek
- Molecular Pharmacology and Physiology, College of Medicine, University of South Florida, Tampa, FL, USA
| | - M N Gordon
- USF Health Byrd Alzheimer's Institute, Tampa, FL 33613, USA
| | - D H Cribbs
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA
| | - N Petrovsky
- Flinders Med. Ctr., Bedford Park, Adelaide 5042, Australia
| | - M G Agadjanyan
- The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA
| | - A Ghochikyan
- The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA
| | - D Morgan
- USF Health Byrd Alzheimer's Institute, Tampa, FL 33613, USA
| |
Collapse
|
148
|
Dhakal S, Kushairi N, Phan CW, Adhikari B, Sabaratnam V, Macreadie I. Dietary Polyphenols: A Multifactorial Strategy to Target Alzheimer's Disease. Int J Mol Sci 2019; 20:E5090. [PMID: 31615073 PMCID: PMC6834216 DOI: 10.3390/ijms20205090] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/11/2019] [Accepted: 10/11/2019] [Indexed: 02/06/2023] Open
Abstract
Ageing is an inevitable fundamental process for people and is their greatest risk factor for neurodegenerative disease. The ageing processes bring changes in cells that can drive the organisms to experience loss of nutrient sensing, disrupted cellular functions, increased oxidative stress, loss of cellular homeostasis, genomic instability, accumulation of misfolded protein, impaired cellular defenses and telomere shortening. Perturbation of these vital cellular processes in neuronal cells can lead to life threatening neurological disorders like Alzheimer's Disease, Parkinson's Disease, Huntington's Disease, Lewy body dementia, etc. Alzheimer's Disease is the most frequent cause of deaths in the elderly population. Various therapeutic molecules have been designed to overcome the social, economic and health care burden caused by Alzheimer's Disease. Almost all the chemical compounds in clinical practice have been found to treat symptoms only limiting them to palliative care. The reason behind such imperfect drugs may result from the inefficiencies of the current drugs to target the cause of the disease. Here, we review the potential role of antioxidant polyphenolic compounds that could possibly be the most effective preventative strategy against Alzheimer's Disease.
Collapse
Affiliation(s)
- Sudip Dhakal
- School of Science, RMIT University, Bundoora, Victoria 3083, Australia.
| | - Naufal Kushairi
- Mushroom Research Centre, University of Malaya, 50603 Kuala Lumpur, Malaysia.
- Department of Anatomy, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Chia Wei Phan
- Mushroom Research Centre, University of Malaya, 50603 Kuala Lumpur, Malaysia.
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Benu Adhikari
- School of Science, RMIT University, Bundoora, Victoria 3083, Australia.
| | - Vikineswary Sabaratnam
- Mushroom Research Centre, University of Malaya, 50603 Kuala Lumpur, Malaysia.
- Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Ian Macreadie
- School of Science, RMIT University, Bundoora, Victoria 3083, Australia.
| |
Collapse
|
149
|
Loera-Valencia R, Cedazo-Minguez A, Kenigsberg PA, Page G, Duarte AI, Giusti P, Zusso M, Robert P, Frisoni GB, Cattaneo A, Zille M, Boltze J, Cartier N, Buee L, Johansson G, Winblad B. Current and emerging avenues for Alzheimer's disease drug targets. J Intern Med 2019; 286:398-437. [PMID: 31286586 DOI: 10.1111/joim.12959] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD), the most frequent cause of dementia, is escalating as a global epidemic, and so far, there is neither cure nor treatment to alter its progression. The most important feature of the disease is neuronal death and loss of cognitive functions, caused probably from several pathological processes in the brain. The main neuropathological features of AD are widely described as amyloid beta (Aβ) plaques and neurofibrillary tangles of the aggregated protein tau, which contribute to the disease. Nevertheless, AD brains suffer from a variety of alterations in function, such as energy metabolism, inflammation and synaptic activity. The latest decades have seen an explosion of genes and molecules that can be employed as targets aiming to improve brain physiology, which can result in preventive strategies for AD. Moreover, therapeutics using these targets can help AD brains to sustain function during the development of AD pathology. Here, we review broadly recent information for potential targets that can modify AD through diverse pharmacological and nonpharmacological approaches including gene therapy. We propose that AD could be tackled not only using combination therapies including Aβ and tau, but also considering insulin and cholesterol metabolism, vascular function, synaptic plasticity, epigenetics, neurovascular junction and blood-brain barrier targets that have been studied recently. We also make a case for the role of gut microbiota in AD. Our hope is to promote the continuing research of diverse targets affecting AD and promote diverse targeting as a near-future strategy.
Collapse
Affiliation(s)
- R Loera-Valencia
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - A Cedazo-Minguez
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | | | - G Page
- Neurovascular Unit and Cognitive impairments - EA3808, University of Poitiers, Poitiers, France
| | - A I Duarte
- CNC- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - P Giusti
- Dipartimento di Scienze del Farmaco, Università degli Studi di Padova, Padova, Italy
| | - M Zusso
- Dipartimento di Scienze del Farmaco, Università degli Studi di Padova, Padova, Italy
| | - P Robert
- CoBTeK - lab, CHU Nice University Côte d'Azur, Nice, France
| | - G B Frisoni
- University Hospitals and University of Geneva, Geneva, Switzerland
| | - A Cattaneo
- University Hospitals and University of Geneva, Geneva, Switzerland
| | - M Zille
- Institute of Experimental and Clinical Pharmacology and Toxicology, Lübeck, Germany
| | - J Boltze
- School of Life Sciences, The University of Warwick, Coventry, UK
| | - N Cartier
- Preclinical research platform, INSERM U1169/MIRCen Commissariat à l'énergie atomique, Fontenay aux Roses, France.,Université Paris-Sud, Orsay, France
| | - L Buee
- Alzheimer & Tauopathies, LabEx DISTALZ, CHU-Lille, Inserm, Univ. Lille, Lille, France
| | - G Johansson
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - B Winblad
- Division of Neurogeriatrics, Centre for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden.,Theme Aging, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
150
|
Fließbach K, McCormick C, Kaulen B, Schneider A. [Anti-tau therapies-what can be expected?]. DER NERVENARZT 2019; 90:891-897. [PMID: 31332452 DOI: 10.1007/s00115-019-0758-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Alzheimer's disease is histopathologically characterized by aggregation of two proteins, namely amyloid-beta peptide and tau protein. Whereas former intervention trials focused particularly on the amyloid pathology, recent therapeutic approaches are directed against the tau pathology. This article summarizes recent progress in anti-tau therapies, especially therapies based on anti-tau immunization and antisense oligonucleotides (ASO).
Collapse
Affiliation(s)
- Klaus Fließbach
- Klinik für Neurodegenerative Erkrankungen und Gerontopsychiatrie, Universitätsklinikum Bonn Venusberg, Campus 1, 53127, Bonn, Deutschland.,Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE, Bonn, Deutschland
| | - Cornelia McCormick
- Klinik für Neurodegenerative Erkrankungen und Gerontopsychiatrie, Universitätsklinikum Bonn Venusberg, Campus 1, 53127, Bonn, Deutschland.,Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE, Bonn, Deutschland
| | - Barbara Kaulen
- Klinik für Neurodegenerative Erkrankungen und Gerontopsychiatrie, Universitätsklinikum Bonn Venusberg, Campus 1, 53127, Bonn, Deutschland.,Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE, Bonn, Deutschland
| | - Anja Schneider
- Klinik für Neurodegenerative Erkrankungen und Gerontopsychiatrie, Universitätsklinikum Bonn Venusberg, Campus 1, 53127, Bonn, Deutschland. .,Deutsches Zentrum für Neurodegenerative Erkrankungen, DZNE, Bonn, Deutschland.
| |
Collapse
|