51
|
Ghosh S, Ali R, Verma S. Aβ-oligomers: A potential therapeutic target for Alzheimer's disease. Int J Biol Macromol 2023; 239:124231. [PMID: 36996958 DOI: 10.1016/j.ijbiomac.2023.124231] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/22/2023] [Accepted: 03/25/2023] [Indexed: 03/30/2023]
Abstract
The cascade of amyloid formation relates to multiple complex events at the molecular level. Previous research has established amyloid plaque deposition as the leading cause of Alzheimer's disease (AD) pathogenesis, detected mainly in aged population. The primary components of the plaques are two alloforms of amyloid-beta (Aβ), Aβ1-42 and Aβ1-40 peptides. Recent studies have provided considerable evidence contrary to the previous claim indicating that amyloid-beta oligomers (AβOs) as the main culprit responsible for AD-associated neurotoxicity and pathogenesis. In this review, we have discussed the primary features of AβOs, such as assembly formation, the kinetics of oligomer formation, interactions with various membranes/membrane receptors, the origin of toxicity, and oligomer-specific detection methods. Recently, the discovery of rationally designed antibodies has opened a gateway for using synthesized peptides as a grafting component in the complementarity determining region (CDR) of antibodies. Thus, the Aβ sequence motif or the complementary peptide sequence in the opposite strand of the β-sheet (extracted from the Protein Data Bank: PDB) helps design oligomer-specific inhibitors. The microscopic event responsible for oligomer formation can be targeted, and thus prevention of the overall macroscopic behaviour of the aggregation or the associated toxicity can be achieved. We have carefully reviewed the oligomer formation kinetics and associated parameters. Besides, we have depicted a thorough understanding of how the synthesized peptide inhibitors can impede the early aggregates (oligomers), mature fibrils, monomers, or a mixture of the species. The oligomer-specific inhibitors (peptides or peptide fragments) lack in-depth chemical kinetics and optimization control-based screening. In the present review, we have proposed a hypothesis for effectively screening oligomer-specific inhibitors using the chemical kinetics (determining the kinetic parameters) and optimization control strategy (cost-dependent analysis). Further, it may be possible to implement the structure-kinetic-activity-relationship (SKAR) strategy instead of structure-activity-relationship (SAR) to improve the inhibitor's activity. The controlled optimization of the kinetic parameters and dose usage will be beneficial for narrowing the search window for the inhibitors.
Collapse
|
52
|
Vogt ACS, Jennings GT, Mohsen MO, Vogel M, Bachmann MF. Alzheimer's Disease: A Brief History of Immunotherapies Targeting Amyloid β. Int J Mol Sci 2023; 24:3895. [PMID: 36835301 PMCID: PMC9961492 DOI: 10.3390/ijms24043895] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/08/2023] [Accepted: 02/11/2023] [Indexed: 02/17/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and may contribute to 60-70% of cases. Worldwide, around 50 million people suffer from dementia and the prediction is that the number will more than triple by 2050, as the population ages. Extracellular protein aggregation and plaque deposition as well as accumulation of intracellular neurofibrillary tangles, all leading to neurodegeneration, are the hallmarks of brains with Alzheimer's disease. Therapeutic strategies including active and passive immunizations have been widely explored in the last two decades. Several compounds have shown promising results in many AD animal models. To date, only symptomatic treatments are available and because of the alarming epidemiological data, novel therapeutic strategies to prevent, mitigate, or delay the onset of AD are required. In this mini-review, we focus on our understanding of AD pathobiology and discuss current active and passive immunomodulating therapies targeting amyloid-β protein.
Collapse
Affiliation(s)
- Anne-Cathrine S. Vogt
- Department of Rheumatology and Immunology (RI), University Hospital, 3010 Bern, Switzerland
- Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, 3008 Bern, Switzerland
| | | | - Mona O. Mohsen
- Department of Rheumatology and Immunology (RI), University Hospital, 3010 Bern, Switzerland
- Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, 3008 Bern, Switzerland
| | - Monique Vogel
- Department of Rheumatology and Immunology (RI), University Hospital, 3010 Bern, Switzerland
- Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, 3008 Bern, Switzerland
| | - Martin F. Bachmann
- Department of Rheumatology and Immunology (RI), University Hospital, 3010 Bern, Switzerland
- Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, 3008 Bern, Switzerland
- Centre for Cellular and Molecular Physiology (CCMP), Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford OX3 7BN, UK
| |
Collapse
|
53
|
Selles MC, Fortuna JTS, Cercato MC, Santos LE, Domett L, Bitencourt ALB, Carraro MF, Souza AS, Janickova H, Azevedo CV, Campos HC, de Souza JM, Alves-Leon S, Prado VF, Prado MAM, Epstein AL, Salvetti A, Longo BM, Arancio O, Klein WL, Sebollela A, De Felice FG, Jerusalinsky DA, Ferreira ST. AAV-mediated neuronal expression of an scFv antibody selective for Aβ oligomers protects synapses and rescues memory in Alzheimer models. Mol Ther 2023; 31:409-419. [PMID: 36369741 PMCID: PMC9931599 DOI: 10.1016/j.ymthe.2022.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 10/25/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022] Open
Abstract
The accumulation of soluble oligomers of the amyloid-β peptide (AβOs) in the brain has been implicated in synapse failure and memory impairment in Alzheimer's disease. Here, we initially show that treatment with NUsc1, a single-chain variable-fragment antibody (scFv) that selectively targets a subpopulation of AβOs and shows minimal reactivity to Aβ monomers and fibrils, prevents the inhibition of long-term potentiation in hippocampal slices and memory impairment induced by AβOs in mice. As a therapeutic approach for intracerebral antibody delivery, we developed an adeno-associated virus vector to drive neuronal expression of NUsc1 (AAV-NUsc1) within the brain. Transduction by AAV-NUsc1 induced NUsc1 expression and secretion in adult human brain slices and inhibited AβO binding to neurons and AβO-induced loss of dendritic spines in primary rat hippocampal cultures. Treatment of mice with AAV-NUsc1 prevented memory impairment induced by AβOs and, remarkably, reversed memory deficits in aged APPswe/PS1ΔE9 Alzheimer's disease model mice. These results support the feasibility of immunotherapy using viral vector-mediated gene delivery of NUsc1 or other AβO-specific single-chain antibodies as a potential therapeutic approach in Alzheimer's disease.
Collapse
Affiliation(s)
- Maria Clara Selles
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; Skirball Institute for Biomolecular Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Juliana T S Fortuna
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-590, Brazil
| | - Magali C Cercato
- Laboratorio de Neuroplasticidad y Neurotoxinas, Instituto de Biología Celular y Neurociencia "Profesor Eduardo De Robertis," Universidad de Buenos Aires/CONICET, Buenos Aires 1121, Argentina
| | - Luis Eduardo Santos
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Luciana Domett
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-590, Brazil
| | - Andre L B Bitencourt
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto 14049-900, Brazil
| | - Mariane Favero Carraro
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto 14049-900, Brazil
| | - Amanda S Souza
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Helena Janickova
- Department of Physiology & Pharmacology and Department of Anatomy & Cell Biology, Robarts Research Institute, The University of Western Ontario, London, ON N6A 5K8, Canada
| | - Caroline Vieira Azevedo
- Laboratório de Neurofisiologia, Departamento de Fisiologia, Universidade Federal de São Paulo, São Paulo 05508-000, Brazil
| | - Henrique Correia Campos
- Laboratório de Neurofisiologia, Departamento de Fisiologia, Universidade Federal de São Paulo, São Paulo 05508-000, Brazil
| | - Jorge M de Souza
- Division of Neurosurgery and Division of Neurology/Epilepsy Program, Clementino Fraga Filho University Hospital, Rio de Janeiro 21941-617, Brazil
| | - Soniza Alves-Leon
- Division of Neurosurgery and Division of Neurology/Epilepsy Program, Clementino Fraga Filho University Hospital, Rio de Janeiro 21941-617, Brazil
| | - Vania F Prado
- Department of Physiology & Pharmacology and Department of Anatomy & Cell Biology, Robarts Research Institute, The University of Western Ontario, London, ON N6A 5K8, Canada
| | - Marco A M Prado
- Department of Physiology & Pharmacology and Department of Anatomy & Cell Biology, Robarts Research Institute, The University of Western Ontario, London, ON N6A 5K8, Canada
| | - Alberto L Epstein
- UMR INSERM U1179-UVSQ, Université de Versailles Saint Quentin en Yvelines, 78180 Montigny-le-Bretonneux, France
| | - Anna Salvetti
- CIRI - Centre International de Recherche en Infectiologie, University of Lyon, Université Claude Bernard Lyon 1, INSERM, U1111, CNRS, UMR5308, ENS Lyon, 69007 Lyon, France
| | - Beatriz Monteiro Longo
- Laboratório de Neurofisiologia, Departamento de Fisiologia, Universidade Federal de São Paulo, São Paulo 05508-000, Brazil
| | - Ottavio Arancio
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY 10032, USA
| | - William L Klein
- Department of Neurobiology, Northwestern University, Evanston, IL 60201, USA
| | - Adriano Sebollela
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto 14049-900, Brazil
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; Centre for Neuroscience Studies, Department of Molecular and Biomedical Sciences & Department of Psychiatry, Queen's University, Kingston, ON K7L 3N6, Canada; D'Or Institute for Research and Education, Rio de Janeiro 22281-100, Brazil
| | - Diana A Jerusalinsky
- Laboratorio de Neuroplasticidad y Neurotoxinas, Instituto de Biología Celular y Neurociencia "Profesor Eduardo De Robertis," Universidad de Buenos Aires/CONICET, Buenos Aires 1121, Argentina
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; D'Or Institute for Research and Education, Rio de Janeiro 22281-100, Brazil; Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-170, Brazil.
| |
Collapse
|
54
|
Chen HY, Zhao Y, Xie YZ. Immunosenescence of brain accelerates Alzheimer's disease progression. Rev Neurosci 2023; 34:85-101. [PMID: 35791032 DOI: 10.1515/revneuro-2022-0021] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/04/2022] [Indexed: 01/07/2023]
Abstract
Most of Alzheimer's disease (AD) cases are sporadic and occur after age 65. With prolonged life expectancy and general population aging, AD is becoming a significant public health concern. The immune system supports brain development, plasticity, and homeostasis, yet it is particularly vulnerable to aging-related changes. Aging of the immune system, called immunosenescence, is the multifaceted remodeling of the immune system during aging. Immunosenescence is a contributing factor to various age-related diseases, including AD. Age-related changes in brain immune cell phenotype and function, crosstalk between immune cells and neural cells, and neuroinflammation work together to promote neurodegeneration and age-related cognitive impairment. Although numerous studies have confirmed the correlation between systemic immune changes and AD, few studies focus on the immune state of brain microenvironment in aging and AD. This review mainly addresses the changes of brain immune microenvironment in aging and AD. Specifically, we delineate how various aspects of the brain immune microenvironment, including immune gateways, immune cells, and molecules, and the interplay between immune cells and neural cells, accelerate AD pathogenesis during aging. We also propose a theoretical framework of therapeutic strategies selectively targeting the different mechanisms to restore brain immune homeostasis.
Collapse
Affiliation(s)
- Hou-Yu Chen
- Department of Abdominal Surgery, Affiliated Cancer Hospital, Institute of Guangzhou Medical University, Guangdong 510095, China
| | - Yan Zhao
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.,Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan 410011, China
| | - Yong-Zhi Xie
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| |
Collapse
|
55
|
Gao S, Wang Y, Ma T, Zhang J. The Late Stage of Abnormal Aging: Dementia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1419:157-167. [PMID: 37418213 DOI: 10.1007/978-981-99-1627-6_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
With the growth of the aging population, more age-related diseases endanger the health of the elderly, and therefore more research attention has been put on Alzheimer's disease and dementia. Dementia does not only posing a serious threat to basic daily living in old age but also impose a greater burden on social and medical care as well as the economy. It is urgent to explore the pathogenesis of Alzheimer's disease and develop effective medicine to prevent or mitigate its onset. Currently, many related mechanisms of the pathogenesis of Alzheimer's disease have been proposed, such as beta-amyloid (A) theory, Tau protein theory, and nerve and blood vessel theory. In addition, from the perspective of improving cognitive function and controlling mental state, dementia-related therapeutic drugs were developed, such as anti-amyloid agents, amyloid vaccine, tau vaccine, and tau-aggregation inhibitor. These theories of pathogenesis and the development of drugs provide valuable experience to lift the veil of cognitive disorders in the future.
Collapse
Affiliation(s)
- Shudan Gao
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
- School of Psychology, Shandong Normal University, Jinan, China
| | - Yun Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
| | - Tao Ma
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Junying Zhang
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
56
|
Feng W, Zhang Y, Sun P, Xiao M. Acquired immunity and Alzheimer's disease. J Biomed Res 2023; 37:15-29. [PMID: 36165328 PMCID: PMC9898041 DOI: 10.7555/jbr.36.20220083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by progressive cognitive defects. The role of the central immune system dominated by microglia in the progression of AD has been extensively investigated. However, little is known about the peripheral immune system in AD pathogenesis. Recently, with the discovery of the meningeal lymphatic vessels and glymphatic system, the roles of the acquired immunity in the maintenance of central homeostasis and neurodegenerative diseases have attracted an increasing attention. The T cells not only regulate the function of neurons, astrocytes, microglia, oligodendrocytes and brain microvascular endothelial cells, but also participate in the clearance of β-amyloid (Aβ) plaques. Apart from producing antibodies to bind Aβ peptides, the B cells affect Aβ-related cascades via a variety of antibody-independent mechanisms. This review systemically summarizes the recent progress in understanding pathophysiological roles of the T cells and B cells in AD.
Collapse
Affiliation(s)
- Weixi Feng
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu 211166, China,Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Chinese Academy of Sciences, Shanghai 200031, China,Weixi Feng, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, Jiangsu 211166, China. Tel: +86-25-86869338; E-mail:
| | - Yanli Zhang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu 211166, China,Brain Institute, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Peng Sun
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu 211166, China,Brain Institute, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu 211166, China,Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Chinese Academy of Sciences, Shanghai 200031, China,Brain Institute, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
57
|
Jung M, Lee S, Park S, Hong J, Kim C, Cho I, Sohn HS, Kim K, Park IW, Yoon S, Kwon S, Shin J, Lee D, Kang M, Go S, Moon S, Chung Y, Kim Y, Kim BS. A Therapeutic Nanovaccine that Generates Anti-Amyloid Antibodies and Amyloid-specific Regulatory T Cells for Alzheimer's Disease. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2207719. [PMID: 36329674 DOI: 10.1002/adma.202207719] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/23/2022] [Indexed: 06/16/2023]
Abstract
Alzheimer's disease (AD), the most common cause of dementia, is a complex condition characterized by multiple pathophysiological mechanisms including amyloid-β (Aβ) plaque accumulation and neuroinflammation in the brain. The current immunotherapy approaches, such as anti-Aβ monoclonal antibody (mAb) therapy, Aβ vaccines, and adoptive regulatory T (Treg) cell transfer, target a single pathophysiological mechanism, which may lead to unsatisfactory therapeutic efficacy. Furthermore, Aβ vaccines often induce T helper 1 (Th1) cell-mediated inflammatory responses. Here, a nanovaccine composed of lipid nanoparticles loaded with Aβ peptides and rapamycin is developed, which targets multiple pathophysiological mechanisms, exhibits the combined effects of anti-Aβ antibody therapy and adoptive Aβ-specific Treg cell transfer, and can overcome the limitations of current immunotherapy approaches for AD. The Nanovaccine effectively delivers rapamycin and Aβ peptides to dendritic cells, produces both anti-Aβ antibodies and Aβ-specific Treg cells, removes Aβ plaques in the brain, alleviates neuroinflammation, prevents Th1 cell-mediated excessive immune responses, and inhibits cognitive impairment in mice. The nanovaccine shows higher efficacy in cognitive recovery than an Aβ vaccine. Unlike anti-Aβ mAb therapy and adoptive Treg cell transfer, both of which require complicated and costly manufacturing processes, the nanovaccine is easy-to-prepare and cost-effective. The nanovaccines can represent a novel treatment option for AD.
Collapse
Affiliation(s)
- Mungyo Jung
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Songmin Lee
- Department of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - Sohui Park
- Department of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - Jihye Hong
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Cheesue Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Illhwan Cho
- Department of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - Hee Su Sohn
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kyunghwan Kim
- Department of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - In Wook Park
- Department of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - Soljee Yoon
- Department of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
- Department of Integrative Biotechnology and Translational Medicine, Yonsei University, Incheon, 21983, Republic of Korea
| | - Sungpil Kwon
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jisu Shin
- Department of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - Donghee Lee
- Department of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
| | - Mikyung Kang
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seokhyung Go
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sangjun Moon
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yeonseok Chung
- Laboratory of Immune Regulation, Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - YoungSoo Kim
- Department of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983, Republic of Korea
- Department of Integrative Biotechnology and Translational Medicine, Yonsei University, Incheon, 21983, Republic of Korea
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Chemical Processes, Institute of Engineering Research, BioMAX, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
58
|
Lambracht-Washington D, Fu M, Manouchehri N, Hynan LS, Stuve O, Rosenberg RN. Glial cell transcriptome analyses in 3xTg-AD mice: Effects of aging, disease progression, and anti-Aβ immunotherapy. AGING BRAIN 2023; 3:100066. [PMID: 36911262 PMCID: PMC9997156 DOI: 10.1016/j.nbas.2023.100066] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 01/02/2023] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
Background To investigate how changes in expression of glial genes relate to a progression of Alzheimer's disease (AD) pathology, and how anti-Aβ immunotherapy impact these changes, we conducted a transcriptomic analysis for brains from cohorts of 2-, 10-, and 20 month old 3xTg-AD mice, and a cross-sectional study in groups of 20 month-old mice treated with active DNA Aβ42 immunization, passive immunotherapy, untreated, and wild-type (wt) controls. Methods Twenty-four Formalin-Fixed Paraffin-Embedded (FFPE) mouse brain sections were used for the gene expression analyses (nanostring). Adjacent sections from these and additional mouse brains were stained for microglia using antibodies detecting IbaI and Gal3. For a semi-quantitative analysis of increased tau and amyloid pathology with aging and disease progression, a comparison of ELISA results from brains of 12 and 20 months old 3xTg-AD mice were shown. Results Based on the different comparisons of transcript numbers found the 3xTg-AD age groups with the senescent 20 months old wt control mouse brains, and the 20 months old 3xTg-AD mouse brains with the 20 months old wt control mouse brains, genes were assigned as upregulated due to aging, or due to disease progression, or due to both. The immunohistochemistry of microglia markers revealed that Gal3 might be an important marker for phagocytosing microglia around amyloid plaques. The comparison of the two anti-Aβ immunotherapy approaches showed a differential downregulation of inflammatory glial genes. Conclusion These results are relevant for future clinical trials using active anti-amyloid immunotherapy.
Collapse
Affiliation(s)
| | - 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
| | - Olaf Stuve
- Department of Neurology, UT Southwestern Medical Center Dallas, USA.,VA North Texas Health Care System, VA Medical Center Dallas, USA
| | | |
Collapse
|
59
|
Rudan Njavro J, Vukicevic M, Fiorini E, Dinkel L, Müller SA, Berghofer A, Bordier C, Kozlov S, Halle A, Buschmann K, Capell A, Giudici C, Willem M, Feederle R, Lichtenthaler SF, Babolin C, Montanari P, Pfeifer A, Kosco-Vilbois M, Tahirovic S. Beneficial Effect of ACI-24 Vaccination on Aβ Plaque Pathology and Microglial Phenotypes in an Amyloidosis Mouse Model. Cells 2022; 12:cells12010079. [PMID: 36611872 PMCID: PMC9818422 DOI: 10.3390/cells12010079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/09/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Amyloid-β (Aβ) deposition is an initiating factor in Alzheimer's disease (AD). Microglia are the brain immune cells that surround and phagocytose Aβ plaques, but their phagocytic capacity declines in AD. This is in agreement with studies that associate AD risk loci with genes regulating the phagocytic function of immune cells. Immunotherapies are currently pursued as strategies against AD and there are increased efforts to understand the role of the immune system in ameliorating AD pathology. Here, we evaluated the effect of the Aβ targeting ACI-24 vaccine in reducing AD pathology in an amyloidosis mouse model. ACI-24 vaccination elicited a robust and sustained antibody response in APPPS1 mice with an accompanying reduction of Aβ plaque load, Aβ plaque-associated ApoE and dystrophic neurites as compared to non-vaccinated controls. Furthermore, an increased number of NLRP3-positive plaque-associated microglia was observed following ACI-24 vaccination. In contrast to this local microglial activation at Aβ plaques, we observed a more ramified morphology of Aβ plaque-distant microglia compared to non-vaccinated controls. Accordingly, bulk transcriptomic analysis revealed a trend towards the reduced expression of several disease-associated microglia (DAM) signatures that is in line with the reduced Aβ plaque load triggered by ACI-24 vaccination. Our study demonstrates that administration of the Aβ targeting vaccine ACI-24 reduces AD pathology, suggesting its use as a safe and cost-effective AD therapeutic intervention.
Collapse
Affiliation(s)
| | | | | | - Lina Dinkel
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Stephan A. Müller
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 80333 Munich, Germany
| | - Anna Berghofer
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 80333 Munich, Germany
| | - Chiara Bordier
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Stanislav Kozlov
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Annett Halle
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Katrin Buschmann
- Biomedical Center (BMC), Ludwig-Maximilians University Munich, 80539 Munich, Germany
| | - Anja Capell
- Biomedical Center (BMC), Ludwig-Maximilians University Munich, 80539 Munich, Germany
| | - Camilla Giudici
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Michael Willem
- Biomedical Center (BMC), Ludwig-Maximilians University Munich, 80539 Munich, Germany
| | - Regina Feederle
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Monoclonal Antibody Core Facility, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), 85764 Neuherberg, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Stefan F. Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 80333 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | | | | | | | | | - Sabina Tahirovic
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Correspondence:
| |
Collapse
|
60
|
Gao L, Shuai Y, Wen L, Zhang H, Zhang Y, Zhang X. Benefit and safety of antibiotics for Alzheimer's disease: Protocol for a systematic review and meta-analysis. Medicine (Baltimore) 2022; 101:e31637. [PMID: 36451430 PMCID: PMC9704868 DOI: 10.1097/md.0000000000031637] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is an age-related degenerative change of the central nervous system, the cause of which remains unclear. Recent studies have found that brain inflammation caused by microbial infections may be one of the etiologies of AD, and antibiotics as novel treatments may be beneficial for delaying the development of AD. Several prospective studies have investigated the effects of different antibiotics on Alzheimer's disease. However, no systematic review or meta-analysis has evaluated the benefits and safety of antibiotics in AD patients. METHODS This study will analyze randomized controlled trials and observational studies published from database inception to December 31, 2022, and included direct or indirect evidence. Studies will be retrieved by searching PubMed, Scopus, Web of Science, Cochrane Central Register of Controlled Clinical Trials, CNKI, and Wan Fang databases. The outcomes of this study included the Alzheimer's Disease Assessment Scale cognitive subscale (ADAS-cog), Montreal Cognitive Assessment (MoCA), Standardized Mini-Mental State Examination (SMMSE), Clinical Dementia Rating (CDR), Frontal Functioning Scale (FAB), Dysfunctional Behavior Rating Instrument (DBRI), Activities of Daily Living (ADLs) Index, and Geriatric Depression Scale (GDS). The risk of bias will be assessed using the Cochrane risk-of-bias assessment instrument for randomized controlled trials. A random-effect/fixed-effects model will be used to summarize the estimates of the mean difference/risk ratio using a 95% confidence interval. RESULTS This study will analyze the benefits and safety of antibiotics in patients with AD. CONCLUSION The results of this analysis will provide evidence to evaluate the benefits and safety of antibiotics in the treatment of AD.
Collapse
Affiliation(s)
- Lin Gao
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Yinqi Shuai
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Lihong Wen
- The Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan Province, China
| | - Hong Zhang
- Chengdu University of Traditional Chinese Medicine Affiliated Hospital, Chengdu, Sichuan Province, China
| | - Yi Zhang
- Chengdu University of Traditional Chinese Medicine Affiliated Hospital, Chengdu, Sichuan Province, China
| | - Xiaoyun Zhang
- Chengdu University of Traditional Chinese Medicine Affiliated Hospital, Chengdu, Sichuan Province, China
- * Correspondence: Xiaoyun Zhang, Chengdu University of Traditional Chinese Medicine Affiliated Hospital, Chengdu, Sichuan Province 610000, China (e-mail: )
| |
Collapse
|
61
|
De Vlieger L, Vandenbroucke RE, Van Hoecke L. Recent insights into viral infections as a trigger and accelerator in alzheimer's disease. Drug Discov Today 2022; 27:103340. [PMID: 35987492 PMCID: PMC9385395 DOI: 10.1016/j.drudis.2022.103340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/08/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder for which only symptomatic medication is available, except for the recently FDA-approved aducanumab. This lack of effective treatment urges us to investigate alternative paths that might contribute to disease development. In light of the recent SARS-CoV-2 pandemic and the disturbing neurological complications seen in some patients, it is desirable to (re)investigate the viability of the viral infection theory claiming that a microbe could affect AD initiation and/or progression. Here, we review the most important evidence for this theory with a special focus on two viruses, namely HSV-1 and SARS-CoV-2. Moreover, we discuss the possible involvement of extracellular vesicles (EVs). This overview will contribute to a more rational approach of potential treatment strategies for AD patients.
Collapse
Affiliation(s)
- Lize De Vlieger
- Barriers in Inflammation Lab, VIB Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Roosmarijn E Vandenbroucke
- Barriers in Inflammation Lab, VIB Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| | - Lien Van Hoecke
- Barriers in Inflammation Lab, VIB Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
62
|
Saleki K, Mohamadi MH, Banazadeh M, Alijanizadeh P, Javanmehr N, Pourahmad R, Nouri HR. In silico design of a TLR4-mediating multiepitope chimeric vaccine against amyotrophic lateral sclerosis via advanced immunoinformatics. J Leukoc Biol 2022; 112:1191-1207. [PMID: 35707959 DOI: 10.1002/jlb.6ma0721-376rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 05/18/2020] [Accepted: 05/25/2020] [Indexed: 12/24/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is the most prevalent motor neuron disorder worldwide. In ALS, progressing disease can result from misfolding and aggregation of superoxide dismutase-1 (SOD1) or TAR DNA-binding protein 43 kDa (TDP43). An efficient immunotherapy for ALS should spare intact SOD1 while eliminating its dysfunctional variant. We utilized advanced immunoinformatics to suggest a potential vaccine candidate against ALS by proposing a model of dynamic TLR4 mediation and induction of a specific Th2-biased shift against mutant SOD1, TDP43, and TRAF6, a protein that specifically interacts with dysfunctional SOD1. SOD1, TDP43, and TRAF6 were retrieved in FASTA. Immune Epitopes Database and CTLpred suggested T/B-cell epitopes from disease-specific regions of selected antigens. A TLR4-mediating adjuvant, RS01, was used. Sequences were assembled via suitable linkers. Tertiary structure of the protein was calculated. Refined protein structure and physicochemical features of the 3D structure were verified in silico. Differential immune induction was assessed via C-ImmSim. GROningen MAchine for Chemical Simulation was used to assess evolution of the docked vaccine-TLR4 complex in blood. Our protein showed high structural quality and was nonallergenic and immune inducing. Also, the vaccine-TLR4 complex stability was verified by RMSD, RMSF, gyration, and visual analyses of the molecular dynamic trajectory. Contact residues in the vaccine-TLR4 complex showed favorable binding energies. Immune stimulation analyses of the proposed candidate demonstrated a sustained memory cell response and a strong adaptive immune reaction. We proposed a potential vaccine candidate against ALS and verified its physicochemical and immune inducing features. Future studies should assess this vaccine in animal studies.
Collapse
Affiliation(s)
- Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran.,USERN Office, Babol University of Medical Sciences, Babol, Iran
| | | | - Mohamad Banazadeh
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Parsa Alijanizadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran.,USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Nima Javanmehr
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran.,USERN Office, Babol University of Medical Sciences, Babol, Iran
| | - Ramtin Pourahmad
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Nouri
- USERN Office, Babol University of Medical Sciences, Babol, Iran.,Immunoregulation Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
63
|
Decourt B, Noorda K, Noorda K, Shi J, Sabbagh MN. Review of Advanced Drug Trials Focusing on the Reduction of Brain Beta-Amyloid to Prevent and Treat Dementia. J Exp Pharmacol 2022; 14:331-352. [PMID: 36339394 PMCID: PMC9632331 DOI: 10.2147/jep.s265626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 10/14/2022] [Indexed: 11/21/2022] Open
Abstract
Alzheimer disease (AD) is the most common neurodegenerative disease and typically affects patients older than age 65. Around this age, the number of neurons begins to gradually decrease in healthy brains, but brains of patients with AD show a marked increase in neuron death, often resulting in a significant loss of cognitive abilities. Cognitive skills affected include information retention, recognition capabilities, and language skills. At present, AD can be definitively diagnosed only through postmortem brain biopsies via the detection of extracellular amyloid beta (Aβ) plaques and intracellular hyperphosphorylated tau neurofibrillary tangles. Because the levels of both Aβ plaques and tau tangles are increased, these 2 proteins are thought to be related to disease progression. Although relatively little is known about the cause of AD and its exact pathobiological development, many forms of treatment have been investigated to determine an effective method for managing AD symptoms by targeting Aβ. These treatments include but are not limited to using small molecules to alter the interactions of Aβ monomers, reducing hyperactivation of neuronal circuits altering Aβ's molecular pathway of synthesis, improving degradation of Aβ, employing passive immunity approaches, and stimulating patients' active immunity to target Aβ. This review summarizes the current therapeutic interventions in Phase II/III of clinical development or higher that are capable of reducing abnormal brain Aβ levels to determine which treatments show the greatest likelihood of clinical efficacy. We conclude that, in the near future, the most promising therapeutic interventions for brain Aβ pathology will likely be passive immunotherapies, with aducanumab and donanemab leading the way, and that these drugs may be combined with antidepressants and acetylcholine esterase inhibitors, which can modulate Aβ synthesis.
Collapse
Affiliation(s)
- Boris Decourt
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | | | | | - Jiong Shi
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Marwan N Sabbagh
- Alzheimer’s and Memory Disorders Division, Department of Neurology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ, USA
| |
Collapse
|
64
|
Kakoti BB, Bezbaruah R, Ahmed N. Therapeutic drug repositioning with special emphasis on neurodegenerative diseases: Threats and issues. Front Pharmacol 2022; 13:1007315. [PMID: 36263141 PMCID: PMC9574100 DOI: 10.3389/fphar.2022.1007315] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 09/12/2022] [Indexed: 11/21/2022] Open
Abstract
Drug repositioning or repurposing is the process of discovering leading-edge indications for authorized or declined/abandoned molecules for use in different diseases. This approach revitalizes the traditional drug discovery method by revealing new therapeutic applications for existing drugs. There are numerous studies available that highlight the triumph of several drugs as repurposed therapeutics. For example, sildenafil to aspirin, thalidomide to adalimumab, and so on. Millions of people worldwide are affected by neurodegenerative diseases. According to a 2021 report, the Alzheimer's disease Association estimates that 6.2 million Americans are detected with Alzheimer's disease. By 2030, approximately 1.2 million people in the United States possibly acquire Parkinson's disease. Drugs that act on a single molecular target benefit people suffering from neurodegenerative diseases. Current pharmacological approaches, on the other hand, are constrained in their capacity to unquestionably alter the course of the disease and provide patients with inadequate and momentary benefits. Drug repositioning-based approaches appear to be very pertinent, expense- and time-reducing strategies for the enhancement of medicinal opportunities for such diseases in the current era. Kinase inhibitors, for example, which were developed for various oncology indications, demonstrated significant neuroprotective effects in neurodegenerative diseases. This review expounds on the classical and recent examples of drug repositioning at various stages of drug development, with a special focus on neurodegenerative disorders and the aspects of threats and issues viz. the regulatory, scientific, and economic aspects.
Collapse
Affiliation(s)
- Bibhuti Bhusan Kakoti
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, India
| | | | | |
Collapse
|
65
|
Downey J, Lam JC, Li VO, Gozes I. Somatic Mutations and Alzheimer’s Disease. J Alzheimers Dis 2022; 90:475-493. [DOI: 10.3233/jad-220643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Alzheimer’s disease (AD) represents a global health challenge, with an estimated 55 million people suffering from the non-curable disease across the world. While amyloid-β plaques and tau neurofibrillary tangles in the brain define AD proteinopathy, it has become evident that diverse coding and non-coding regions of the genome may significantly contribute to AD neurodegeneration. The diversity of factors associated with AD pathogenesis, coupled with age-associated damage, suggests that a series of triggering events may be required to initiate AD. Since somatic mutations accumulate with aging, and aging is a major risk factor for AD, there is a great potential for somatic mutational events to drive disease. Indeed, recent data from the Gozes team/laboratories as well as other leading laboratories correlated the accumulation of somatic brain mutations with the progression of tauopathy. In this review, we lay the current perspectives on the principal genetic factors associated with AD and the potential causes, highlighting the contribution of somatic mutations to the pathogenesis of late onset Alzheimer’s disease. The roles that artificial intelligence and big data can play in accelerating the progress of causal somatic mutation markers/biomarkers identification, and the associated drug discovery/repurposing, have been highlighted for future AD and other neurodegenerative studies, with the aim to bring hope for the vulnerable aging population.
Collapse
Affiliation(s)
- Jocelyn Downey
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
| | - Jacqueline C.K. Lam
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
- Department of Computer Science and Technology, University of Cambridge, UK
| | - Victor O.K. Li
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China
| | - Illana Gozes
- The Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Adams Super Center for Brain Studies and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
66
|
Knecht L, Folke J, Dodel R, Ross JA, Albus A. Alpha-synuclein Immunization Strategies for Synucleinopathies in Clinical Studies: A Biological Perspective. Neurotherapeutics 2022; 19:1489-1502. [PMID: 36083395 PMCID: PMC9606184 DOI: 10.1007/s13311-022-01288-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2022] [Indexed: 11/29/2022] Open
Abstract
The therapeutic strategies currently available for neurodegenerative diseases such as Parkinson's disease target only the symptoms of the disease. Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy can be summarized as synucleinopathies, as they are all characterized by the aggregation and accumulation of alpha-synuclein (α-syn) in the brain. Targeting α-syn by its formation and progression opens a new and promising disease-modifying therapeutic strategy. Thus, several distinct immunotherapeutic approaches are currently being evaluated in clinical trials. The objective of this article is to review, from a biological perspective, the most important properties of these passive and active immunotherapies to point out their relevance and suitability for the treatment of synucleinopathies.
Collapse
Affiliation(s)
- Luisa Knecht
- Chair of Geriatric Medicine, University Duisburg-Essen, Essen, Germany
| | - Jonas Folke
- Chair of Geriatric Medicine, University Duisburg-Essen, Essen, Germany
- Centre for Neuroscience & Stereology, Department of Neurology, Bispebjerg-Frederiksberg Hospital, University Hospital of Copenhagen, 2400, Copenhagen, Denmark
| | - Richard Dodel
- Chair of Geriatric Medicine, University Duisburg-Essen, Essen, Germany.
| | - J Alexander Ross
- Chair of Geriatric Medicine, University Duisburg-Essen, Essen, Germany
| | - Alexandra Albus
- Chair of Geriatric Medicine, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
67
|
Chen Y, Colonna M. Spontaneous and induced adaptive immune responses in Alzheimer's disease: new insights into old observations. Curr Opin Immunol 2022; 77:102233. [PMID: 35839620 DOI: 10.1016/j.coi.2022.102233] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/28/2022] [Accepted: 06/06/2022] [Indexed: 01/20/2023]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the elderly. Although AD is primarily a neurological disorder distinguished by amyloid β plaques and intracellular neurofibrillary tangles, the immune system can impact the progression of the disease and may be targeted for therapeutic purposes. To date, most studies have focused on innate immune responses of microglia. However, emerging evidence implicates adaptive immune responses by T cells and B cells in the progression of AD. Moreover, the recent approval of an antibody that promotes amyloid β plaque clearance for AD therapy has pinpointed adaptive immunity as a fertile ground for the design of novel therapeutic approaches. Here, we highlight key studies delineating T cell and B cell responses in human AD and mouse models of AD, identify open questions on the specificity, development and impact of these responses and discuss outlooks for future studies and novel therapeutic avenues.
Collapse
Affiliation(s)
- Yun Chen
- Department of Pathology and Immunology and Department of Neurology, Washington University School of Medicine in St Louis, USA
| | - Marco Colonna
- Department of Pathology and Immunology and Department of Neurology, Washington University School of Medicine in St Louis, USA.
| |
Collapse
|
68
|
Smith KJ, Gwyer Findlay E. Expression of antimicrobial host defence peptides in the central nervous system during health and disease. DISCOVERY IMMUNOLOGY 2022; 1:kyac003. [PMID: 38566904 PMCID: PMC10917193 DOI: 10.1093/discim/kyac003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/13/2022] [Accepted: 07/21/2022] [Indexed: 04/04/2024]
Abstract
Antimicrobial host defence peptides (HDP) are critical for the first line of defence against bacterial, viral, and fungal pathogens. Over the past decade we have become more aware that, in addition to their antimicrobial roles, they also possess the potent immunomodulatory capacity. This includes chemoattracting immune cells, activating dendritic cells and macrophages, and altering T-cell differentiation. Most examinations of their immunomodulatory roles have focused on tissues in which they are very abundant, such as the intestine and the inflamed skin. However, HDP have now been detected in the brain and the spinal cord during a number of conditions. We propose that their presence in the central nervous system (CNS) during homeostasis, infection, and neurodegenerative disease has the potential to contribute to immunosurveillance, alter host responses and skew developing immunity. Here, we review the evidence for HDP expression and function in the CNS in health and disease. We describe how a wide range of HDP are expressed in the CNS of humans, rodents, birds, and fish, suggesting a conserved role in protecting the brain from pathogens, with evidence of production by resident CNS cells. We highlight differences in methodology used and how this may have resulted in the immunomodulatory roles of HDP being overlooked. Finally, we discuss what HDP expression may mean for CNS immune responses.
Collapse
Affiliation(s)
- Katie J Smith
- Centre for Inflammation Research, University of Edinburgh, 47 Little France Crescent, EH16 4TJ, Edinburgh, UK
| | - Emily Gwyer Findlay
- Centre for Inflammation Research, University of Edinburgh, 47 Little France Crescent, EH16 4TJ, Edinburgh, UK
| |
Collapse
|
69
|
Sidoroff V, Bower P, Stefanova N, Fanciulli A, Stankovic I, Poewe W, Seppi K, Wenning GK, Krismer F. Disease-Modifying Therapies for Multiple System Atrophy: Where Are We in 2022? JOURNAL OF PARKINSON'S DISEASE 2022; 12:1369-1387. [PMID: 35491799 PMCID: PMC9398078 DOI: 10.3233/jpd-223183] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Multiple system atrophy is a rapidly progressive and fatal neurodegenerative disorder. While numerous preclinical studies suggested efficacy of potentially disease modifying agents, none of those were proven to be effective in large-scale clinical trials. Three major strategies are currently pursued in preclinical and clinical studies attempting to slow down disease progression. These target α-synuclein, neuroinflammation, and restoration of neurotrophic support. This review provides a comprehensive overview on ongoing preclinical and clinical developments of disease modifying therapies. Furthermore, we will focus on potential shortcomings of previous studies that can be avoided to improve data quality in future studies of this rare disease.
Collapse
Affiliation(s)
- Victoria Sidoroff
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Pam Bower
- The Multiple System Atrophy Coalition, Inc., McLean, VA, USA
| | - Nadia Stefanova
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Iva Stankovic
- Neurology Clinic, University Clinical Center of Serbia, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Werner Poewe
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Klaus Seppi
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Gregor K Wenning
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Krismer
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
70
|
Plowey ED, Bussiere T, Rajagovindan R, Sebalusky J, Hamann S, von Hehn C, Castrillo-Viguera C, Sandrock A, Budd Haeberlein S, van Dyck CH, Huttner A. Alzheimer disease neuropathology in a patient previously treated with aducanumab. Acta Neuropathol 2022; 144:143-153. [PMID: 35581440 PMCID: PMC9217863 DOI: 10.1007/s00401-022-02433-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 05/05/2022] [Accepted: 05/05/2022] [Indexed: 12/13/2022]
Abstract
Amyloid beta (Aβ) plaque is a defining pathologic feature of Alzheimer disease (AD). Aducanumab, a monoclonal IgG1 that selectively binds aggregated species of Aβ, has been shown by amyloid positron emission tomography (Amyloid PET) to reduce Aβ plaques in patients with prodromal and mild AD. This is the first autopsy report of the AD neuropathology in a patient previously treated with aducanumab. The patient was an 84-year-old woman who was randomized to the placebo arm of the PRIME Phase 1b study (221AD103). The patient progressed to moderate dementia (MMSE = 14/30), beyond the targeted early AD treatment stage, before receiving aducanumab in the long-term extension (LTE). The patient then received 32 monthly doses of aducanumab, titrated up to 6 mg/kg, for a cumulative dose of 186 mg/kg. In the LTE, Amyloid PET scans demonstrated robust Aβ plaque reduction, from a composite standard uptake value ratio (SUVR) of 1.5 at screening to < 1.1 at 56 weeks post-aducanumab dosing. MRI examinations were negative for amyloid-related imaging abnormalities (ARIA). She passed away in hospice care 4 months after her last dose of aducanumab. The postmortem neuropathologic examination confirmed AD neuropathologic changes. Aβ and IBA1 immunohistochemistry assays demonstrated sparse residual Aβ plaque engaged by amoeboid reactive microglia. Phospho-Tau (pTau) immunohistochemistry demonstrated neocortical neurofibrillary degeneration (Braak stage V, NIA/AA Stage B3). However, the density of pTau neuropathology, including neuritic plaque pTau (NP-Tau), appeared lower in the PRIME LTE Patient compared to a reference cohort of untreated Braak stage V-VI, NIA/AA Stage B3 AD cases. Taken together, this case report is the first to provide Amyloid PET and neuropathologic evidence substantiating the impact of aducanumab to reduce Aβ plaque neuropathology in a patient with AD. Furthermore, this report underscores the critical importance of autopsy neuropathology studies to augment our understanding of aducanumab's mechanism of action and impact on AD biomarkers.
Collapse
Affiliation(s)
- Edward D Plowey
- Research and Development, Biogen, 225 Binney Street, Cambridge, MA, 02142, USA.
| | - Thierry Bussiere
- Research and Development, Biogen, 225 Binney Street, Cambridge, MA, 02142, USA
| | - Raj Rajagovindan
- Research and Development, Biogen, 225 Binney Street, Cambridge, MA, 02142, USA
| | - Jennifer Sebalusky
- Research and Development, Biogen, 225 Binney Street, Cambridge, MA, 02142, USA
| | - Stefan Hamann
- Research and Development, Biogen, 225 Binney Street, Cambridge, MA, 02142, USA
| | - Christian von Hehn
- Research and Development, Biogen, 225 Binney Street, Cambridge, MA, 02142, USA
| | | | - Alfred Sandrock
- Research and Development, Biogen, 225 Binney Street, Cambridge, MA, 02142, USA
| | | | | | - Anita Huttner
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
71
|
Grimmer T. [Therapy Developments in Alzheimer's Disease]. FORTSCHRITTE DER NEUROLOGIE-PSYCHIATRIE 2022; 90:352-360. [PMID: 35588740 DOI: 10.1055/a-1802-4837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The development of new therapies to treat Alzheimer's disease is a focus of global drug discovery. Research is being conducted into more potent therapies for symptomatic treatment, particularly for behavioral disturbances, but also into drugs that intervene in the pathophysiology of the disease, with the aim of halting or at least slowing the progression of the disease. To this end, the focus of identifying people with Alzheimer's disease is shifting to stages of pre-dementia such as that of Mild Cognitive Impairment (MCI), almost synonymous with prodromal AD, or even to asymptomatic stages. Currently, passive immunization using monoclonal antibodies against Aβ42 has shown the most encouraging results. However, it has not been possible to demonstrate statistically significant differences on the primary target parameters in multiple completed pivotal trials. The anti-amyloid antibody aducanumab received conditional preliminary approval in the U.S. based on amyloid reduction; approval for its use in Europe is an ongoing process. Current pharmacological treatments of Alzheimer's disease offer limited symptomatic benefit. No drugs to delay progression of the disease is yet on the market in Germany. Therefore, it is recommended that patients, especially those in pre-dementia stages or at the onset of Alzheimer's disease, be encouraged to participate in clinical trials in order to help develop new drugs that are more effective in the treatment of this disease and that can then benefit many more patients in the future.
Collapse
Affiliation(s)
- Timo Grimmer
- Psychiatry and Psychotherapy, Klinikum rechts der Isar der Technischen Universität München, München, Germany
| |
Collapse
|
72
|
Jeong H, Shin H, Hong S, Kim Y. Physiological Roles of Monomeric Amyloid-β and Implications for Alzheimer's Disease Therapeutics. Exp Neurobiol 2022; 31:65-88. [PMID: 35673997 PMCID: PMC9194638 DOI: 10.5607/en22004] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/26/2022] [Accepted: 03/30/2022] [Indexed: 12/23/2022] Open
Abstract
Alzheimer's disease (AD) progressively inflicts impairment of synaptic functions with notable deposition of amyloid-β (Aβ) as senile plaques within the extracellular space of the brain. Accordingly, therapeutic directions for AD have focused on clearing Aβ plaques or preventing amyloidogenesis based on the amyloid cascade hypothesis. However, the emerging evidence suggests that Aβ serves biological roles, which include suppressing microbial infections, regulating synaptic plasticity, promoting recovery after brain injury, sealing leaks in the blood-brain barrier, and possibly inhibiting the proliferation of cancer cells. More importantly, these functions were found in in vitro and in vivo investigations in a hormetic manner, that is to be neuroprotective at low concentrations and pathological at high concentrations. We herein summarize the physiological roles of monomeric Aβ and current Aβ-directed therapies in clinical trials. Based on the evidence, we propose that novel therapeutics targeting Aβ should selectively target Aβ in neurotoxic forms such as oligomers while retaining monomeric Aβ in order to preserve the physiological functions of Aβ monomers.
Collapse
Affiliation(s)
- Hyomin Jeong
- Division of Integrated Science and Engineering, Underwood International College, Yonsei University, Incheon 21983, Korea
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Korea
| | - Heewon Shin
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Korea
| | - Seungpyo Hong
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Yonsei Frontier Lab, Yonsei University, Seoul 03722, Korea
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - YoungSoo Kim
- Division of Integrated Science and Engineering, Underwood International College, Yonsei University, Incheon 21983, Korea
- Department of Pharmacy, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Korea
- Yonsei Frontier Lab, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
73
|
Ho G, Choo PC, Waragai M, Inoue S, Masliah E, Hashimoto M. Reconsideration of Alzheimer's Disease Therapy from a Viewpoint of Amyloidogenic Evolvability. J Alzheimers Dis Rep 2022; 6:207-210. [PMID: 35591950 PMCID: PMC9108623 DOI: 10.3233/adr-210021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 04/03/2022] [Indexed: 11/26/2022] Open
Abstract
Presuming that Alzheimer's disease (AD) might represent an antagonistic pleiotropic phenomenon derived from the evolvability of multiple amyloidogenic proteins, targeting such proteins simultaneously could enhance therapeutic efficacy. Furthermore, considering that amyloid-β (Aβ) immunotherapies during reproductive life stage might adversely decrease Aβ evolvability in an offspring's brain, the disease-modifying Aβ immunotherapies should be limited to post-reproductive time in lifespan. Thus, current Aβ immunotherapy strategies should be revised accordingly. Given that the "adiponectin paradox" might underlie both amyloidosis and cognitive dysfunction in aging brain, blocking activin signaling situated downstream of the adiponectin paradox might be an alternative strategy to prevent AD.
Collapse
Affiliation(s)
- Gilbert Ho
- PCND Neuroscience Research Institute, Poway, CA, USA
| | - Pei Chen Choo
- PCND Neuroscience Research Institute, Poway, CA, USA
| | - Masaaki Waragai
- Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Satoshi Inoue
- Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute of Gerontology, Itabashi-ku, Tokyo, Japan
- Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Eliezer Masliah
- Division of Neuroscience, National Institute on Aging, Bethesda, MD, USA
| | - Makoto Hashimoto
- Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| |
Collapse
|
74
|
Villain N. Therapeutic news in Alzheimer's disease: Soon a disease-modifying therapy? Rev Neurol (Paris) 2022; 178:437-440. [PMID: 35489824 DOI: 10.1016/j.neurol.2022.02.456] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 02/28/2022] [Indexed: 11/18/2022]
Abstract
Research on disease-modifying treatments for Alzheimer's disease has resulted in a series of failures over the past 20 years. However, in the last three years, four molecules have shown significant effects on clinical endpoints in phase II or III clinical trials (i.e., slowing of cognitive decline). Among these four molecules, three are anti-amyloid immunotherapies: aducanumab, donanemab, and lecanemab, responsible for a significant clearance of cerebral beta-amyloid deposits. These provisional data are still awaiting confirmation to put an end to the controversy surrounding the 2021 Food and Drug Administration's decision to give conditional approval to aducanumab, which is considered premature by many specialists. Confirmation is also necessary to assess the benefit (magnitude of the slowing of cognitive decline) and risk (edema and cerebral hemorrhage induced by these treatments) balance of these molecules. Masitinib, a treatment whose probable mechanism of action is neuroinflammation, has also shown positive effects that need to be confirmed. Therapies targeting the tau protein are less advanced and have yet to be proven. Patients have renewed hope since it may not be unreasonable that these disease-modifying therapies will be part of the French therapeutic arsenal within the next five years.
Collapse
Affiliation(s)
- N Villain
- AP-HP Sorbonne Université, Hôpital Pitié-Salpêtrière, Département de Neurologie, Institut de la Mémoire et de la Maladie d'Alzheimer, 47-83, boulevard de l'Hôpital, 75651 Paris Cedex 13, France; Institut du Cerveau - ICM, Sorbonne Université, INSERM U1127, CNRS 7225, Paris, France.
| |
Collapse
|
75
|
Alves SS, da Silva Junior RMP, Delfino-Pereira P, Pereira MGAG, Vasconcelos I, Schwaemmle H, Mazzei RF, Carlos ML, Espreafico EM, Tedesco AC, Sebollela A, Almeida SS, de Oliveira JAC, Garcia-Cairasco N. A Genetic Model of Epilepsy with a Partial Alzheimer's Disease-Like Phenotype and Central Insulin Resistance. Mol Neurobiol 2022; 59:3721-3737. [PMID: 35378696 DOI: 10.1007/s12035-022-02810-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/22/2022] [Indexed: 12/20/2022]
Abstract
Studies have suggested an important connection between epilepsy and Alzheimer's disease (AD), mostly due to the high number of patients diagnosed with AD who develop epileptic seizures later on. However, this link is not well understood. Previous studies from our group have identified memory impairment and metabolic abnormalities in the Wistar audiogenic rat (WAR) strain, a genetic model of epilepsy. Our goal was to investigate AD behavioral and molecular alterations, including brain insulin resistance, in naïve (seizure-free) animals of the WAR strain. We used the Morris water maze (MWM) test to evaluate spatial learning and memory performance and hippocampal tissue to verify possible molecular and immunohistochemical alterations. WARs presented worse performance in the MWM test (p < 0.0001), higher levels of hyperphosphorylated tau (S396) (p < 0.0001) and phosphorylated glycogen synthase kinase 3 (S21/9) (p < 0.05), and lower insulin receptor levels (p < 0.05). Conversely, WARs and Wistar controls present progressive increase in amyloid fibrils (p < 0.0001) and low levels of soluble amyloid-β. Interestingly, the detected alterations were age-dependent, reaching larger differences in aged than in young adult animals. In summary, the present study provides evidence of a partial AD-like phenotype, including altered regulation of insulin signaling, in a genetic model of epilepsy. Together, these data contribute to the understanding of the connection between epilepsy and AD as comorbidities. Moreover, since both tau hyperphosphorylation and altered insulin signaling have already been reported in epilepsy and AD, these two events should be considered as important components in the interconnection between epilepsy and AD pathogenesis and, therefore, potential therapeutic targets in this field.
Collapse
Affiliation(s)
- Suélen Santos Alves
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirao Preto, Brazil
| | | | - Polianna Delfino-Pereira
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirao Preto, Brazil
| | | | - Israel Vasconcelos
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirao Preto, Brazil
| | - Hanna Schwaemmle
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirao Preto, Brazil
| | - Rodrigo Focosi Mazzei
- Department of Psychology, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo (FFCLRP-USP), Ribeirao Preto, Brazil
| | - Maiko Luiz Carlos
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo (FFCLRP-USP), Ribeirao Preto, Brazil
| | - Enilza Maria Espreafico
- Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirao Preto, Brazil
| | - Antônio Claudio Tedesco
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo (FFCLRP-USP), Ribeirao Preto, Brazil
| | - Adriano Sebollela
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirao Preto, Brazil
| | - Sebastião Sousa Almeida
- Department of Psychology, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo (FFCLRP-USP), Ribeirao Preto, Brazil
| | - José Antônio Cortes de Oliveira
- Department of Physiology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Av. Dos Bandeirantes 3900, Ribeirao Preto, SP, 14049-900, Brazil
| | - Norberto Garcia-Cairasco
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirao Preto, Brazil.
- Department of Physiology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Av. Dos Bandeirantes 3900, Ribeirao Preto, SP, 14049-900, Brazil.
| |
Collapse
|
76
|
Primary angiitis of central nervous system – A challenging diagnosis. J Neuroimmunol 2022; 366:577844. [DOI: 10.1016/j.jneuroim.2022.577844] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/26/2022] [Accepted: 03/06/2022] [Indexed: 12/14/2022]
|
77
|
Xie J, Van Hoecke L, Vandenbroucke RE. The Impact of Systemic Inflammation on Alzheimer's Disease Pathology. Front Immunol 2022; 12:796867. [PMID: 35069578 PMCID: PMC8770958 DOI: 10.3389/fimmu.2021.796867] [Citation(s) in RCA: 142] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is a devastating age-related neurodegenerative disorder with an alarming increasing prevalence. Except for the recently FDA-approved Aducanumab of which the therapeutic effect is not yet conclusively proven, only symptomatic medication that is effective for some AD patients is available. In order to be able to design more rational and effective treatments, our understanding of the mechanisms behind the pathogenesis and progression of AD urgently needs to be improved. Over the last years, it became increasingly clear that peripheral inflammation is one of the detrimental factors that can contribute to the disease. Here, we discuss the current understanding of how systemic and intestinal (referred to as the gut-brain axis) inflammatory processes may affect brain pathology, with a specific focus on AD. Moreover, we give a comprehensive overview of the different preclinical as well as clinical studies that link peripheral Inflammation to AD initiation and progression. Altogether, this review broadens our understanding of the mechanisms behind AD pathology and may help in the rational design of further research aiming to identify novel therapeutic targets.
Collapse
Affiliation(s)
- Junhua Xie
- VIB Center for Inflammation Research, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Lien Van Hoecke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Roosmarijn E Vandenbroucke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
78
|
Developing an Effective Peptide-Based Vaccine for COVID-19: Preliminary Studies in Mice Models. Viruses 2022; 14:v14030449. [PMID: 35336856 PMCID: PMC8954996 DOI: 10.3390/v14030449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/12/2022] [Accepted: 02/21/2022] [Indexed: 02/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) has caused massive health and economic disasters worldwide. Although several vaccines have effectively slowed the spread of the virus, their long-term protection and effectiveness against viral variants are still uncertain. To address these potential shortcomings, this study proposes a peptide-based vaccine to prevent COVID-19. A total of 15 B cell epitopes of the wild-type severe acute respiratory coronavirus 2 (SARS-CoV-2) spike (S) protein were selected, and their HLA affinities predicted in silico. Peptides were divided into two groups and tested in C57BL/6 mice with either QS21 or Al(OH)3 as the adjuvant. Our results demonstrated that the peptide-based vaccine stimulated high and durable antibody responses in mice, with the T and B cell responses differing based on the type of adjuvant employed. Using epitope mapping, we showed that our peptide-based vaccine produced antibody patterns similar to those in COVID-19 convalescent individuals. Moreover, plasma from vaccinated mice and recovered COVID-19 humans had the same neutralizing activity when tested with a pseudo particle assay. Our data indicate that this adjuvant peptide-based vaccine can generate sustainable and effective B and T cell responses. Thus, we believe that our peptide-based vaccine can be a safe and effective vaccine against COVID-19, particularly because of the flexibility of including new peptides to prevent emerging SARS-CoV-2 variants and avoiding unwanted autoimmune responses.
Collapse
|
79
|
Wang L, Wei X. T Cell-Mediated Autoimmunity in Glaucoma Neurodegeneration. Front Immunol 2022; 12:803485. [PMID: 34975917 PMCID: PMC8716691 DOI: 10.3389/fimmu.2021.803485] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/01/2021] [Indexed: 02/05/2023] Open
Abstract
Glaucoma as the leading neurodegenerative disease leads to blindness in 3.6 million people aged 50 years and older worldwide. For many decades, glaucoma therapy has primarily focused on controlling intraocular pressure (IOP) and sound evidence supports its role in delaying the progress of retinal ganglial cell (RGC) damage and protecting patients from vision loss. Meanwhile, accumulating data point to the immune-mediated attack of the neural retina as the underlying pathological process behind glaucoma that may come independent of raised IOP. Recently, some scholars have suggested autoimmune aspects in glaucoma, with autoreactive T cells mediating the chief pathogenic process. This autoimmune process, as well as the pathological features of glaucoma, largely overlaps with other neurodegenerative diseases in the central nervous system (CNS), including Alzheimer’s disease, Parkinson’s disease, and multiple sclerosis. In addition, immune modulation therapy, which is regarded as a potential solution for glaucoma, has been boosted in trials in some CNS neurodegenerative diseases. Thus, novel insights into the T cell-mediated immunity and treatment in CNS neurodegenerative diseases may serve as valuable inspirations for ophthalmologists. This review focuses on the role of T cell-mediated immunity in the pathogenesis of glaucoma and discusses potential applications of relevant findings of CNS neurodegenerative diseases in future glaucoma research.
Collapse
Affiliation(s)
- Lixiang Wang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Wei
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China.,Department of Ophthalmology, Shangjin Nanfu Hospital, Chengdu, China
| |
Collapse
|
80
|
Machine learning prediction and tau-based screening identifies potential Alzheimer's disease genes relevant to immunity. Commun Biol 2022; 5:125. [PMID: 35149761 PMCID: PMC8837797 DOI: 10.1038/s42003-022-03068-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 01/21/2022] [Indexed: 12/19/2022] Open
Abstract
With increased research funding for Alzheimer's disease (AD) and related disorders across the globe, large amounts of data are being generated. Several studies employed machine learning methods to understand the ever-growing omics data to enhance early diagnosis, map complex disease networks, or uncover potential drug targets. We describe results based on a Target Central Resource Database protein knowledge graph and evidence paths transformed into vectors by metapath matching. We extracted features between specific genes and diseases, then trained and optimized our model using XGBoost, termed MPxgb(AD). To determine our MPxgb(AD) prediction performance, we examined the top twenty predicted genes through an experimental screening pipeline. Our analysis identified potential AD risk genes: FRRS1, CTRAM, SCGB3A1, FAM92B/CIBAR2, and TMEFF2. FRRS1 and FAM92B are considered dark genes, while CTRAM, SCGB3A1, and TMEFF2 are connected to TREM2-TYROBP, IL-1β-TNFα, and MTOR-APP AD-risk nodes, suggesting relevance to the pathogenesis of AD.
Collapse
|
81
|
Scherlek AA, Kozberg MG, Nicoll JAR, Perosa V, Freeze WM, van der Weerd L, Bacskai BJ, Greenberg SM, Frosch MP, Boche D, van Veluw SJ. Histopathological correlates of haemorrhagic lesions on ex vivo magnetic resonance imaging in immunized Alzheimer's disease cases. Brain Commun 2022; 4:fcac021. [PMID: 35224489 PMCID: PMC8870423 DOI: 10.1093/braincomms/fcac021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/31/2021] [Accepted: 02/01/2022] [Indexed: 12/14/2022] Open
Abstract
Haemorrhagic amyloid-related imaging abnormalities on MRI are frequently observed adverse events in the context of amyloid β immunotherapy trials in patients with Alzheimer's disease. The underlying histopathology and pathophysiological mechanisms of haemorrhagic amyloid-related imaging abnormalities remain largely unknown, although coexisting cerebral amyloid angiopathy may play a key role. Here, we used ex vivo MRI in cases that underwent amyloid β immunotherapy during life to screen for haemorrhagic lesions and assess underlying tissue and vascular alterations. We hypothesized that these lesions would be associated with severe cerebral amyloid angiopathy. Ten cases were selected from the long-term follow-up study of patients who enrolled in the first clinical trial of active amyloid β immunization with AN1792 for Alzheimer's disease. Eleven matched non-immunized Alzheimer's disease cases from an independent brain brank were used as 'controls'. Formalin-fixed occipital brain slices were imaged at 7 T MRI to screen for haemorrhagic lesions (i.e. microbleeds and cortical superficial siderosis). Samples with and without haemorrhagic lesions were cut and stained. Artificial intelligence-assisted quantification of amyloid β plaque area, cortical and leptomeningeal cerebral amyloid angiopathy area, the density of iron and calcium positive cells and reactive astrocytes and activated microglia was performed. On ex vivo MRI, cortical superficial siderosis was observed in 5/10 immunized Alzheimer's disease cases compared with 1/11 control Alzheimer's disease cases (κ = 0.5). On histopathology, these areas revealed iron and calcium positive deposits in the cortex. Within the immunized Alzheimer's disease group, areas with siderosis on MRI revealed greater leptomeningeal cerebral amyloid angiopathy and concentric splitting of the vessel walls compared with areas without siderosis. Moreover, greater density of iron-positive cells in the cortex was associated with lower amyloid β plaque area and a trend towards increased post-vaccination antibody titres. This work highlights the use of ex vivo MRI to investigate the neuropathological correlates of haemorrhagic lesions observed in the context of amyloid β immunotherapy. These findings suggest a possible role for cerebral amyloid angiopathy in the formation of haemorrhagic amyloid-related imaging abnormalities, awaiting confirmation in future studies that include brain tissue of patients who received passive immunotherapy against amyloid β with available in vivo MRI during life.
Collapse
Affiliation(s)
- Ashley A. Scherlek
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Mariel G. Kozberg
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA,J. Philip Kistler Stroke Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - James A. R. Nicoll
- Clinical Neurosciences, Clinical and Experimental Sciences School, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Valentina Perosa
- J. Philip Kistler Stroke Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Whitney M. Freeze
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Louise van der Weerd
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands,Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Brian J. Bacskai
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Steven M. Greenberg
- J. Philip Kistler Stroke Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Matthew P. Frosch
- Neuropathology Service, C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences School, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Susanne J. van Veluw
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA,J. Philip Kistler Stroke Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA,Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands,Correspondence to: Susanne J. van Veluw MassGeneral Institute for Neurodegenerative Disease Massachusetts General Hospital 114 16th Street Charlestown, 02129 MA, USA E-mail:
| |
Collapse
|
82
|
Rickenbach C, Gericke C. Specificity of Adaptive Immune Responses in Central Nervous System Health, Aging and Diseases. Front Neurosci 2022; 15:806260. [PMID: 35126045 PMCID: PMC8812614 DOI: 10.3389/fnins.2021.806260] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/29/2021] [Indexed: 12/25/2022] Open
Abstract
The field of neuroimmunology endorses the involvement of the adaptive immune system in central nervous system (CNS) health, disease, and aging. While immune cell trafficking into the CNS is highly regulated, small numbers of antigen-experienced lymphocytes can still enter the cerebrospinal fluid (CSF)-filled compartments for regular immune surveillance under homeostatic conditions. Meningeal lymphatics facilitate drainage of brain-derived antigens from the CSF to deep cervical lymph nodes to prime potential adaptive immune responses. During aging and CNS disorders, brain barriers and meningeal lymphatic functions are impaired, and immune cell trafficking and antigen efflux are altered. In this context, alterations in the immune cell repertoire of blood and CSF and T and B cells primed against CNS-derived autoantigens have been observed in various CNS disorders. However, for many diseases, a causal relationship between observed immune responses and neuropathological findings is lacking. Here, we review recent discoveries about the association between the adaptive immune system and CNS disorders such as autoimmune neuroinflammatory and neurodegenerative diseases. We focus on the current challenges in identifying specific T cell epitopes in CNS diseases and discuss the potential implications for future diagnostic and treatment options.
Collapse
Affiliation(s)
- Chiara Rickenbach
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland
| | - Christoph Gericke
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland
| |
Collapse
|
83
|
Yokota H, Hayashi H, Hanaguri J, Yamagami S, Kushiyama A, Nakagami H, Nagaoka T. Effect of prorenin peptide vaccine on the early phase of diabetic retinopathy in a murine model of type 2 diabetes. PLoS One 2022; 17:e0262568. [PMID: 35041699 PMCID: PMC8765632 DOI: 10.1371/journal.pone.0262568] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 12/29/2021] [Indexed: 11/18/2022] Open
Abstract
Prorenin is viewed as an ideal target molecule in the prevention of diabetic retinopathy. However, no drugs are available for inhibiting activation of prorenin. Here, we tested the effect of a prorenin peptide vaccine (VP) in the retina of a murine model of type 2 diabetes (T2D). To choose the optimal vaccine, we selected three different epitopes of the prorenin prosegment (E1, E2, and E3) and conjugated them to keyhole limpet hemocyanin (KLH). We injected C57BL/6J mice twice with KLH only (as a control vaccine), E1 conjugated with KLH (E1-KLH), E2-KLH, or E3-KLH and compared antibody titers. E2-KLH showed the highest antibody titer and specific immunoreactivity of anti-sera against prorenin, so we used E2-KLH as VP. Then, we administered injections to the non-diabetic db/m and diabetic db/db mice, as follows: db/m + KLH, db/db + KLH, and db/db + VP. Retinal blood flow measurement with laser speckle flowgraphy showed that the impaired retinal circulation response to both flicker light and systemic hyperoxia in db/db mice improved with VP. Furthermore, the prolonged implicit time of b-wave and oscillatory potentials in electroretinography was prevented, and immunohistochemical analysis showed reduced microglial activation, gliosis, and vascular leakage. The enzyme-linked immunosorbent spot assay confirmed vaccinated mice had no auto-immune response against prorenin itself. The present data suggest that vaccination against prorenin is an effective and safe measure against the early pathological changes of diabetic retinopathy in T2D.
Collapse
Affiliation(s)
- Harumasa Yokota
- Division of Ophthalmology, Department of Visual Science, Nihon University School of Medicine, Tokyo, Japan
- * E-mail:
| | - Hiroki Hayashi
- Department of Health Development and Medicine, Osaka University, Osaka, Japan
| | - Junya Hanaguri
- Division of Ophthalmology, Department of Visual Science, Nihon University School of Medicine, Tokyo, Japan
| | - Satoru Yamagami
- Division of Ophthalmology, Department of Visual Science, Nihon University School of Medicine, Tokyo, Japan
| | - Akifumi Kushiyama
- Department of Pharmacotherapy, Meiji Pharmaceutical University, Tokyo, Japan
| | - Hironori Nakagami
- Department of Health Development and Medicine, Osaka University, Osaka, Japan
| | - Taiji Nagaoka
- Division of Ophthalmology, Department of Visual Science, Nihon University School of Medicine, Tokyo, Japan
| |
Collapse
|
84
|
Fighting fire with fire: the immune system might be key in our fight against Alzheimer's disease. Drug Discov Today 2022; 27:1261-1283. [PMID: 35032668 DOI: 10.1016/j.drudis.2022.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/25/2021] [Accepted: 01/06/2022] [Indexed: 12/13/2022]
Abstract
The ultimate cause of Alzheimer's disease (AD) is still unknown and no disease-modifying treatment exists. Emerging evidence supports the concept that the immune system has a key role in AD pathogenesis. This awareness leads to the idea that specific parts of the immune system must be engaged to ward off the disease. Immunotherapy has dramatically improved the management of several previously untreatable cancers and could hold similar promise as a novel therapy for treating AD. However, before potent immunotherapies can be rationally designed as treatment against AD, we need to fully understand the dynamic interplay between AD and the different parts of our immune system. Accordingly, here we review the most important aspects of both the innate and adaptive immune system in relation to AD pathology. Teaser: Emerging results support the concept that Alzheimer's disease is affected by the inability of the immune system to contain the pathology of the brain. Here, we discuss how we can engage our immune system to fight this devastating disease.
Collapse
|
85
|
Prachayasittikul V, Pingaew R, Prachayasittikul S, Prachayasittikul V. 8-Hydroxyquinolines: A Promising Pharmacophore Potentially Developed as Disease-Modifying Agents for Neurodegenerative Diseases: A Review. HETEROCYCLES 2022. [DOI: 10.3987/rev-22-sr(r)6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
86
|
Vukicevic M, Fiorini E, Siegert S, Carpintero R, Rincon-Restrepo M, Lopez-Deber P, Piot N, Ayer M, Rentero I, Babolin C, Bravo-Veyrat S, Giriens V, Morici C, Beuzelin M, Gesbert A, Rivot S, Depretti S, Donati P, Streffer J, Pfeifer A, Kosco-Vilbois MH. OUP accepted manuscript. Brain Commun 2022; 4:fcac022. [PMID: 35479516 PMCID: PMC9037369 DOI: 10.1093/braincomms/fcac022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 11/22/2021] [Accepted: 02/01/2022] [Indexed: 11/14/2022] Open
Abstract
Pyroglutamate amyloid beta3–42 (pGlu-Abeta3–42), a highly
amyloidogenic and neurotoxic form of Abeta, is N-terminally truncated to form a
pyroglutamate and has recently been proposed as a key target for immunotherapy.
Optimized ACI-24, a vaccine in development for the treatment and prevention of
Alzheimer’s disease, focuses the antibody response on the first 15
N-terminal amino acids of Abeta (Abeta1–15). Importantly, clinical data
with an initial version of ACI-24 incorporating Abeta1–15, established
the vaccine’s safety and tolerability with evidence of immunogenicity. To
explore optimized ACI-24’s capacity to generate antibodies to
pGlu-Abeta3–42, pre-clinical studies were carried out. Vaccinating mice
and non-human primates demonstrated that optimized ACI-24 was well-tolerated and
induced an antibody response against Abeta1–42 as expected, as well as
high titres of IgG reactive with pyroGlu-Abeta. Epitope mapping of the
polyclonal response confirmed these findings revealing broad coverage of
epitopes particularly for Abeta peptides mimicking where cleavage occurs to form
pGlu-Abeta3–42. These data are in striking contrast to results obtained
with other clinically tested Abeta targeting vaccines which generated restricted
and limited antibody diversity. Taken together, our findings demonstrate that
optimized ACI-24 vaccination represents a breakthrough to provide a safe immune
response with a broader Abeta sequence recognition compared to previously tested
vaccines, creating binders to pathogenic forms of Abeta important in
pathogenesis including pGlu-Abeta3–42.
Collapse
Affiliation(s)
| | | | | | | | | | | | - N. Piot
- AC Immune SA, Lausanne, Switzerland
| | - M. Ayer
- AC Immune SA, Lausanne, Switzerland
| | | | | | | | | | | | | | | | - S. Rivot
- AC Immune SA, Lausanne, Switzerland
| | | | | | - J. Streffer
- AC Immune SA, Lausanne, Switzerland
- Biomedical Sciences, University of
Antwerp, Antwerp, Belgium
| | | | - M. H. Kosco-Vilbois
- AC Immune SA, Lausanne, Switzerland
- Correspondence to: M. Kosco-Vilbois AC Immune, AC Immune
SA EPFL Innovation Park, Building B CH-1015 Lausanne, Switzerland E-mail:
| |
Collapse
|
87
|
Montoliu-Gaya L, Villegas S. Production of Therapeutic Single-Chain Variable Fragments (ScFv) in Pichia pastoris. Methods Mol Biol 2022; 2313:151-167. [PMID: 34478136 DOI: 10.1007/978-1-0716-1450-1_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The interest in the use of monoclonal antibodies as therapeutic molecules has raised in the recent years. Due to their high affinity and specificity towards other biological molecules, antibodies are being widely used to treat a broad range of human diseases such as cancer, rheumatism, and cardiovascular diseases. Currently, the production of IgG-like antibodies is mainly obtained from stable or transient mammalian expression systems that allow proper folding and posttranslational modifications. Despite the technological advances of the last decade, the use of these systems still has a rather high production cost and long processing times. For these reasons, researchers are increasingly interested in alternative antibody production methods as well as alternative antibody formats. Bacterial systems, such as Escherichia coli, are extensively being used for recombinant protein production because their easy manipulation and cheap costs. However, the presence of lipopolysaccharides (LPS) traces in the already fractionated recombinant protein makes these systems not good candidates for the preparation of therapeutic molecules. Yeast systems, such as Pichia pastoris, present the convenient easy manipulation of microbial systems but show some key advantages of eukaryotic expression systems, like improved folding machinery and absence of LPS. They are especially suitable for the production of antibody fragments, which do not need human-like glycosylation, avoiding the high costs of mammalian systems. Here, the protocol for the expression and purification of a single-chain antibody fragment (scFv) in P. pastoris is provided, in deep detail for lab manipulation and briefly for a 5L-bioreactor production.
Collapse
Affiliation(s)
- Laia Montoliu-Gaya
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Sandra Villegas
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular. Facultat de Biociències, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
88
|
Sveikata L, Charidimou A, Viswanathan A. Vessels Sing Their ARIAs: The Role of Vascular Amyloid in the Age of Aducanumab. Stroke 2021; 53:298-302. [PMID: 34905943 DOI: 10.1161/strokeaha.121.036873] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We review the implications of the recently approved aducanumab amyloid-β immunotherapy for treating Alzheimer disease with comorbid cerebral amyloid angiopathy. In clinical trials, amyloid-β immunotherapy has been associated with a high rate of amyloid-related imaging abnormalities, potentially driven by coexisting cerebral amyloid angiopathy. Therefore, immunotherapy's efficacy in patients may be modified by coexisting cerebrovascular pathology. We discuss the contributions of cerebral amyloid angiopathy on the development of amyloid-related imaging abnormalities and propose strategies to identify cerebral amyloid angiopathy in patients considered for immunotherapy.
Collapse
Affiliation(s)
- Lukas Sveikata
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston (L.S., A.C., A.V.).,Department of Clinical Neurosciences, Division of Neurology, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Switzerland (L.S.)
| | - Andreas Charidimou
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston (L.S., A.C., A.V.)
| | - Anand Viswanathan
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston (L.S., A.C., A.V.)
| |
Collapse
|
89
|
Yoo TJ. Anti-Inflammatory Gene Therapy Improves Spatial Memory Performance in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2021; 85:1001-1008. [PMID: 34897091 PMCID: PMC8925118 DOI: 10.3233/jad-215270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The immune system plays a critical role in neurodegenerative processes involved in Alzheimer’s disease (AD). In this study, a gene-based immunotherapeutic method examined the effects of anti-inflammatory cellular immune response elements (CIREs) in the amyloid-β protein precursor (AβPP) mouse model. Bi-monthly intramuscular administration, beginning at either 4 or 6 months, and examined at 7.5 through 16 months, with plasmids encoding Interleukin (IL)-10, IL-4, TGF-β polynucleotides, or a combination thereof, into AβPP mice improved spatial memory performance. This work demonstrates an efficient gene therapy strategy to downregulate neuroinflammation, and possibly prevent or delay cognitive decline in AD.
Collapse
Affiliation(s)
- Tai June Yoo
- Korea Allergy Clinic, KangNam Gu, Seoul, South Korea.,University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
90
|
Nimmo JT, Kelly L, Verma A, Carare RO, Nicoll JAR, Dodart JC. Amyloid-β and α-Synuclein Immunotherapy: From Experimental Studies to Clinical Trials. Front Neurosci 2021; 15:733857. [PMID: 34539340 PMCID: PMC8441015 DOI: 10.3389/fnins.2021.733857] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/13/2021] [Indexed: 11/28/2022] Open
Abstract
Alzheimer’s disease and Lewy body diseases are the most common causes of neurodegeneration and dementia. Amyloid-beta (Aβ) and alpha-synuclein (αSyn) are two key proteins involved in the pathogenesis of these neurodegenerative diseases. Immunotherapy aims to reduce the harmful effects of protein accumulation by neutralising toxic species and facilitating their removal. The results of the first immunisation trial against Aβ led to a small percentage of meningoencephalitis cases which revolutionised vaccine design, causing a shift in the field of immunotherapy from active to passive immunisation. While the vast majority of immunotherapies have been developed for Aβ and tested in Alzheimer’s disease, the field has progressed to targeting other proteins including αSyn. Despite showing some remarkable results in animal models, immunotherapies have largely failed final stages of clinical trials to date, with the exception of Aducanumab recently licenced in the US by the FDA. Neuropathological findings translate quite effectively from animal models to human trials, however, cognitive and functional outcome measures do not. The apparent lack of translation of experimental studies to clinical trials suggests that we are not obtaining a full representation of the effects of immunotherapies from animal studies. Here we provide a background understanding to the key concepts and challenges involved in therapeutic design. This review further provides a comprehensive comparison between experimental and clinical studies in Aβ and αSyn immunotherapy and aims to determine the possible reasons for the disconnection in their outcomes.
Collapse
Affiliation(s)
- Jacqui Taryn Nimmo
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Louise Kelly
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Ajay Verma
- Yumanity Therapeutics, Boston, MA, United States
| | - Roxana O Carare
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - James A R Nicoll
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | | |
Collapse
|
91
|
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder that currently has no cure. The aged population is growing globally, creating an urgent need for more promising therapies for this debilitating disease. Much effort has been made in recent decades, and the field is highly dynamic, with numerous trials. The main focus of these trials includes disease modification and symptomatic treatment. Some have shown beneficial outcomes, while others have shown no significant benefits. Here, we cover the outcome of recently published AD clinical trials, as well as the mechanism of action of these therapeutical agents, to re-think drug development strategies and directions for future studies.
Collapse
|
92
|
Chee SEJ, Solito E. The Impact of Ageing on the CNS Immune Response in Alzheimer's Disease. Front Immunol 2021; 12:738511. [PMID: 34603320 PMCID: PMC8484764 DOI: 10.3389/fimmu.2021.738511] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/01/2021] [Indexed: 12/25/2022] Open
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disease strongly associated with increasing age. Neuroinflammation and the accumulation of amyloid protein are amongst the hallmarks of this disease and most translational research to date has focused on targeting these two processes. However, the exact etiology of AD remains to be fully elucidated. When compared alongside, the immune response in AD closely resembles the central nervous system (CNS) immune changes seen in elderly individuals. It is possible that AD is a pathological consequence of an aged immune system secondary to chronic stimulation by a previous or ongoing insult. Pathological changes like amyloid accumulation and neuronal cell death may reflect this process of immunosenescence as the CNS immune system fails to maintain homeostasis in the CNS. It is likely that future treatments designed to modulate the aged immune system may prove beneficial in altering the disease course. The development of new tests for appropriate biomarkers would also be essential in screening for patients most likely to benefit from such treatments.
Collapse
Affiliation(s)
- Stephan En Jie Chee
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Egle Solito
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Federico II University, Naples, Italy
| |
Collapse
|
93
|
Xiong Y, Lim CS. Understanding the Modulatory Effects of Cannabidiol on Alzheimer's Disease. Brain Sci 2021; 11:brainsci11091211. [PMID: 34573232 PMCID: PMC8472755 DOI: 10.3390/brainsci11091211] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/03/2021] [Accepted: 09/04/2021] [Indexed: 11/23/2022] Open
Abstract
Alzheimer’s disease (AD), the most common neurodegenerative disease, is characterized by progressive cognitive impairment. The deposition of amyloid beta (Aβ) and hyperphosphorylated tau is considered the hallmark of AD pathology. Many therapeutic approaches such as Food and Drug Administration-approved cholinesterase inhibitors and N–methyl–D–aspartate receptor antagonists have been used to intervene in AD pathology. However, current therapies only provide limited symptomatic relief and are ineffective in preventing AD progression. Cannabidiol (CBD), a phytocannabinoid devoid of psychoactive responses, provides neuroprotective effects through both cannabinoid and noncannabinoid receptors. Recent studies using an AD mouse model have suggested that CBD can reverse cognitive deficits along with Aβ-induced neuroinflammatory, oxidative responses, and neuronal death. Furthermore, CBD can reduce the accumulation of Aβ and hyperphosphorylation of tau, suggesting the possibility of delaying AD progression. Particularly, the noncannabinoid receptor, peroxisome proliferator-activated receptor gamma, has been suggested to be involved in multiple functions of CBD. Therefore, understanding the underlying mechanisms of CBD is necessary for intervening in AD pathology in depth and for the translation of preclinical studies into clinical settings. In this review, we summarize recent studies on the effect of CBD in AD and suggest problems to be overcome for the therapeutic use of CBD.
Collapse
Affiliation(s)
- Yinyi Xiong
- Department of Pharmacology, Wonkwang University School of Medicine, Iksan 54538, Korea;
- Department of Rehabilitation, Affiliated Hospital of Jiujiang University, Jiujiang 332000, China
| | - Chae-Seok Lim
- Department of Pharmacology, Wonkwang University School of Medicine, Iksan 54538, Korea;
- Correspondence: ; Tel.: +82-63-850-6765; Fax: +82-63-850-7262
| |
Collapse
|
94
|
Song G, Yang H, Shen N, Pham P, Brown B, Lin X, Hong Y, Sinu P, Cai J, Li X, Leon M, Gordon MN, Morgan D, Zhang S, Cao C. An Immunomodulatory Therapeutic Vaccine Targeting Oligomeric Amyloid-β. J Alzheimers Dis 2021; 77:1639-1653. [PMID: 32925044 DOI: 10.3233/jad-200413] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Aging is considered the most important risk factor for Alzheimer's disease (AD). Recent research supports the theory that immunotherapy targeting the "oligomeric" forms of amyloid-β (Aβ) may halt the progression of AD. However, previous clinical trial of the vaccine against Aβ, called AN1792, was suspended due to cases of meningoencephalitis in patients. OBJECTIVE To develop a peptide sensitized dendritic cells (DCs) vaccine that would target oligomer Aβ and prevent an autoimmune response. METHODS Double transgenic APPswe/PS1ΔE9 (Tg) and C57BL/6J control mice were used in this study. Cytokine expression profile detection, characterization of antisera, brain GSK-3β, LC3 expression, and spatial working memory testing before and post-vaccination were obtained. RESULTS Epitope prediction indicated that E22W42 could generate 13 new T cell epitopes which can strengthen immunity in aged subjects and silence several T cell epitopes of the wild type Aβ. The silenced T cell epitope could help avoid the autoimmune response that was seen in some patients of the AN-1792 vaccine. The E22W42 not only helped sensitize bone marrow-derived DCs for the development of an oligomeric Aβ-specific antibody, but also delayed memory impairment in the APP/PS1 mouse model. Most importantly, this E22W42 peptide will not alter the DC's natural immunomodulatory properties. CONCLUSION The E22W42 vaccine is possibly safer for patients with impaired immune systems. Since there is increasing evidence that oligomeric form of Aβ are the toxic species to neurons, the E22W42 antibody's specificity for these "oligomeric" Aβ species could provide the opportunity to produce some clinical benefits in AD subjects.
Collapse
Affiliation(s)
- Ge Song
- Tianjin University of Traditional Chinese Medicine, Tianjin, P.R. China
| | - Haiqiang Yang
- College of Arts and Science, University of South Florida, Tampa, FL, USA
| | - Ning Shen
- College of Arts and Science, University of South Florida, Tampa, FL, USA
| | - Phillip Pham
- Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Breanna Brown
- Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Xiaoyang Lin
- Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Yuzhu Hong
- Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Paul Sinu
- Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Jianfeng Cai
- College of Arts and Science, University of South Florida, Tampa, FL, USA
| | - Xiaopeng Li
- College of Arts and Science, University of South Florida, Tampa, FL, USA
| | - Michael Leon
- Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
| | - Marcia N Gordon
- Department of Translational Neuroscience, College of Medicine, Michigan State University, Grand Rapids, MI, USA
| | - David Morgan
- Department of Translational Neuroscience, College of Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Sai Zhang
- Institution of Brain Trauma and Neurology Disease, Key Laboratory of Neurotrauma Repair of Tianjin, Tianjin, China
| | - Chuanhai Cao
- College of Arts and Science, University of South Florida, Tampa, FL, USA.,Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA.,College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
95
|
Mahdiabadi S, Momtazmanesh S, Perry G, Rezaei N. Immune modulations and immunotherapies for Alzheimer's disease: a comprehensive review. Rev Neurosci 2021; 33:365-381. [PMID: 34506700 DOI: 10.1515/revneuro-2021-0092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 08/18/2021] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD), the most common cause of dementia, is characterized by progressive cognitive and memory impairment ensued from neuronal dysfunction and eventual death. Intraneuronal deposition of tau proteins and extracellular senile amyloid-β plaques have ruled as the supreme postulations of AD for a relatively long time, and accordingly, a wide range of therapeutics, especially immunotherapies have been implemented. However, none of them resulted in significant positive cognitive outcomes. Especially, the repetitive failure of anti-amyloid therapies proves the inefficiency of the amyloid cascade hypothesis, suggesting that it is time to reconsider this hypothesis. Thus, for the time being, the focus is being shifted to neuroinflammation as a third core pathology in AD. Neuroinflammation was previously considered a result of the two aforementioned phenomena, but new studies suggest that it might play a causal role in the pathogenesis of AD. Neuroinflammation can act as a double-edged sword in the pathogenesis of AD, and the activation of glial cells is indispensable for mediating such attenuating or detrimental effects. The association of immune-related genes polymorphisms with the clinical phenotype of AD as well as the protective effect of anti-inflammatory drugs like nonsteroidal anti-inflammatory drugs supports the possible causal role of neuroinflammation in AD. Here, we comprehensively review immune-based therapeutic approaches toward AD, including monoclonal antibodies and vaccines. We also discuss their efficacy and underlying reasons for shortcomings. Lastly, we highlight the capacity of modulating the neuroimmune interactions and targeting neuroinflammation as a promising opportunity for finding optimal treatments for AD.
Collapse
Affiliation(s)
- Sara Mahdiabadi
- School of Medicine, Tehran University of Medical Sciences, Tehran 1416753955, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Children's Medical Center, Tehran 1419733151, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran 14194, Iran
| | - Sara Momtazmanesh
- School of Medicine, Tehran University of Medical Sciences, Tehran 1416753955, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Children's Medical Center, Tehran 1419733151, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran 14194, Iran
| | - George Perry
- Department of Biology and Neurosciences Institute, University of Texas at San Antonio (UTSA), San Antonio, TX 78249, USA
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Children's Medical Center, Tehran 1419733151, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran 14194, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1416753955, Iran
| |
Collapse
|
96
|
Illouz T, Madar R, Hirsh T, Biragyn A, Okun E. Induction of an effective anti-Amyloid-β humoral response in aged mice. Vaccine 2021; 39:4817-4829. [PMID: 34294479 DOI: 10.1016/j.vaccine.2021.07.023] [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] [Received: 01/21/2021] [Revised: 06/20/2021] [Accepted: 07/08/2021] [Indexed: 01/03/2023]
Abstract
Aging-related decline in immune functions, termed immunosenescence, is a primary cause of reduced protective responses to vaccines in the elderly, due to impaired induction of cellular and humoral responses to new antigens (Ag), especially if the response is T cell dependent. The result is a more severe morbidity following infections, more prolonged and frequent hospitalization, and a higher mortality rate than in the general population. Therefore, there is an increasing need to develop vaccination strategies that overcome immunosenescence, especially for aging-related diseases such as Alzheimer's disease (AD). Here we report a new vaccination strategy harnessing memory-based immunity, which is less affected by aging. We found that aged C57BL/6 and 5xFAD mice exhibit a dramatic reduction in anti-Amyloid-β (Aβ) antibody (Ab) production. We aimed to reverse this process by inducing memory response at a young age. To this end, young mice were primed with the vaccine carrier Hepatitis B surface antigen (HBsAg). At an advanced age, these mice were immunized with an Aβ1-11 fused to HBsAg. This vaccination scheme elicited a markedly higher Aβ-specific antibody titer than vaccinating aged unprimed mice with the same construct. Importantly, this vaccine strategy more efficiently reduced cerebral Aβ levels and altered microglial phenotype. Overall, we provide evidence that priming with an exogenous Ag carrier can overcome impaired humoral responses to self-antigens in the elderly, paving the route for a potent immunotherapy to AD.
Collapse
Affiliation(s)
- Tomer Illouz
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Ravit Madar
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Tamir Hirsh
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Arya Biragyn
- Immunoregulation Section, Laboratory of Molecular Biology and Immunology, NIA, NIH, MD 21224, USA
| | - Eitan Okun
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan 5290002, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan 5290002, Israel.
| |
Collapse
|
97
|
Ojha R, Prajapati VK. Cognizance of posttranslational modifications in vaccines: A way to enhanced immunogenicity. J Cell Physiol 2021; 236:8020-8034. [PMID: 34170014 PMCID: PMC8427110 DOI: 10.1002/jcp.30483] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/21/2021] [Accepted: 06/14/2021] [Indexed: 12/25/2022]
Abstract
Vaccination is a significant advancement or preventative strategy for controlling the spread of various severe infectious and noninfectious diseases. The purpose of vaccination is to stimulate or activate the immune system by injecting antigens, i.e., either whole microorganisms or using the pathogen's antigenic part or macromolecules. Over time, researchers have made tremendous efforts to reduce vaccine side effects or failure by developing different strategies combining with immunoinformatic and molecular biology. These newly designed vaccines are composed of single or several antigenic molecules derived from a pathogenic organism. Although, whole‐cell vaccines are still in use against various diseases but due to their ineffectiveness, other vaccines like DNA‐based, RNA‐based, and protein‐based vaccines, with the addition of immunostimulatory agents, are in the limelight. Despite this, many researchers escape the most common fundamental phenomenon of protein posttranslational modifications during the development of vaccines, which regulates protein functional behavior, evokes immunogenicity and stability, etc. The negligence about post translational modification (PTM) during vaccine development may affect the vaccine's efficacy and immune responses. Therefore, it becomes imperative to consider these modifications of macromolecules before finalizing the antigenic vaccine construct. Here, we have discussed different types of posttranslational/transcriptional modifications that are usually considered during vaccine construct designing: Glycosylation, Acetylation, Sulfation, Methylation, Amidation, SUMOylation, Ubiquitylation, Lipidation, Formylation, and Phosphorylation. Based on the available research information, we firmly believe that considering these modifications will generate a potential and highly immunogenic antigenic molecule against communicable and noncommunicable diseases compared to the unmodified macromolecules.
Collapse
Affiliation(s)
- Rupal Ojha
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| |
Collapse
|
98
|
Waragai M, Ho G, Takamatsu Y, Wada R, Sugama S, Takenouchi T, Masliah E, Hashimoto M. Adiponectin Paradox as a Therapeutic Target in Alzheimer's Disease. J Alzheimers Dis 2021; 76:1249-1253. [PMID: 32623396 PMCID: PMC7504987 DOI: 10.3233/jad-200416] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Despite the apparent neurotoxicity of amyloid-β (Aβ), recent clinical trials of Aβ immunotherapy have not shown any clinical benefit in Alzheimer’s disease (AD). Given this, clarification of the next generation therapeutic strategy in AD is warranted. Hypothetically, adiponectin might be involved in promoting amyloidogenic evolvability in reproduction, which may result in the adiponectin paradox through antagonistic pleiotropy mechanism in aging, leading to AD. Accordingly, preventing the adiponectin paradox by suppressing adiponectin signaling might prove therapeutic in AD.
Collapse
Affiliation(s)
- Masaaki Waragai
- Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Gilbert Ho
- PCND Neuroscience Research Institute, Poway, CA, USA
| | - Yoshiki Takamatsu
- Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Ryoko Wada
- Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Shuei Sugama
- Department of Physiology, Nippon Medical School, Tokyo, Japan
| | - Takato Takenouchi
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Eliezer Masliah
- Division of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Makoto Hashimoto
- Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| |
Collapse
|
99
|
Kozberg MG, Perosa V, Gurol ME, van Veluw SJ. A practical approach to the management of cerebral amyloid angiopathy. Int J Stroke 2021; 16:356-369. [PMID: 33252026 PMCID: PMC9097498 DOI: 10.1177/1747493020974464] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cerebral amyloid angiopathy is a common small vessel disease in the elderly involving vascular amyloid-β deposition. Cerebral amyloid angiopathy is one of the leading causes of intracerebral hemorrhage and a significant contributor to age-related cognitive decline. The awareness of a diagnosis of cerebral amyloid angiopathy is important in clinical practice as it impacts decisions to use lifelong anticoagulation or nonpharmacological alternatives to anticoagulation such as left atrial appendage closure in patients who have concurrent atrial fibrillation, another common condition in older adults. This review summarizes the latest literature regarding the management of patients with sporadic cerebral amyloid angiopathy, including diagnostic criteria, imaging biomarkers for cerebral amyloid angiopathy severity, and management strategies to decrease intracerebral hemorrhage risk. In a minority of patients, the presence of cerebral amyloid angiopathy triggers an autoimmune inflammatory reaction, referred to as cerebral amyloid angiopathy-related inflammation, which is often responsive to immunosuppressive treatment in the acute phase. Diagnosis and management of cerebral amyloid angiopathy-related inflammation will be presented separately. While there are currently no effective therapeutics available to cure or halt the progression of cerebral amyloid angiopathy, we discuss emerging avenues for potential future interventions.
Collapse
Affiliation(s)
- Mariel G Kozberg
- MassGeneral Institute for Neurodegenerative Disease, 2348Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA,USA
| | - Valentina Perosa
- MassGeneral Institute for Neurodegenerative Disease, 2348Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA,USA
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
| | - M Edip Gurol
- Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA,USA
| | - Susanne J van Veluw
- MassGeneral Institute for Neurodegenerative Disease, 2348Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA,USA
| |
Collapse
|
100
|
Alves SS, Silva-Junior RMPD, Servilha-Menezes G, Homolak J, Šalković-Petrišić M, Garcia-Cairasco N. Insulin Resistance as a Common Link Between Current Alzheimer's Disease Hypotheses. J Alzheimers Dis 2021; 82:71-105. [PMID: 34024838 DOI: 10.3233/jad-210234] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Almost 115 years ago, Alois Alzheimer described Alzheimer's disease (AD) for the first time. Since then, many hypotheses have been proposed. However, AD remains a severe health public problem. The current medical approaches for AD are limited to symptomatic interventions and the complexity of this disease has led to a failure rate of approximately 99.6%in AD clinical trials. In fact, no new drug has been approved for AD treatment since 2003. These failures indicate that we are failing in mimicking this disease in experimental models. Although most studies have focused on the amyloid cascade hypothesis of AD, the literature has made clear that AD is rather a multifactorial disorder. Therefore, the persistence in a single theory has resulted in lost opportunities. In this review, we aim to present the striking points of the long scientific path followed since the description of the first AD case and the main AD hypotheses discussed over the last decades. We also propose insulin resistance as a common link between many other hypotheses.
Collapse
Affiliation(s)
- Suélen Santos Alves
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| | - Rui Milton Patrício da Silva-Junior
- Department of Internal Medicine, Ribeirão Preto Medical School -University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil.,Department of Physiology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| | - Gabriel Servilha-Menezes
- Department of Physiology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| | - Jan Homolak
- Department of Pharmacology, University of Zagreb School of Medicine, Zagreb, Croatia.,Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Melita Šalković-Petrišić
- Department of Pharmacology, University of Zagreb School of Medicine, Zagreb, Croatia.,Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Norberto Garcia-Cairasco
- Department of Neurosciences and Behavioral Sciences, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil.,Department of Physiology, Ribeirão Preto Medical School - University of São Paulo (FMRP-USP), Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|