1
|
Liu N, Liang X, Chen Y, Xie L. Recent trends in treatment strategies for Alzheimer 's disease and the challenges: A topical advancement. Ageing Res Rev 2024; 94:102199. [PMID: 38232903 DOI: 10.1016/j.arr.2024.102199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/12/2024] [Indexed: 01/19/2024]
Abstract
Alzheimer's Disease (AD) is an irreversible and progressive neurological disease that has affected at least 50 million people around the globe. Considering the severity of the disease and the continuous increase in the number of patients, the development of new effective drugs or intervention strategies for AD has become urgent. AD is caused by a combination of genetic, environmental, and lifestyle factors, but its exact cause has not yet been clarified. Given the current challenges being faced in the clinical treatment of AD, such as complex AD pathological network and insufficient early diagnosis, herein, we have focused on the three core pathological features of AD, including amyloid-β (Aβ) aggregation, tau phosphorylation and tangles, and activation of inflammatory factors. In this review, we have briefly underscored the primary evidence supporting each pathology and discuss AD pathological network among Aβ, tau, and inflammation. We have also comprehensively summarized the most instructive drugs and their treatment strategies against Aβ, tau, or neuroinflammation used in basic research and clinical trials. Finally, we have discussed and outlined the pros and cons of each pathological approach and looked forward to potential personalized diagnosis and treatment strategies that are beneficial to AD patients.
Collapse
Affiliation(s)
- Ni Liu
- College of Public Health, Zhengzhou University, Zhengzhou 450000, China.
| | - Xiaohan Liang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Yu Chen
- College of Public Health, Zhengzhou University, Zhengzhou 450000, China.
| | - Lihang Xie
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
2
|
Trouche SG, Boutajangout A, Asuni A, Fontés P, Sigurdsson EM, Verdier JM, Mestre-Francés N. Amyloid-β targeting immunisation in aged non-human primate (Microcebus murinus). Brain Behav Immun 2023; 109:63-77. [PMID: 36592872 PMCID: PMC10023341 DOI: 10.1016/j.bbi.2022.12.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/06/2022] [Accepted: 12/29/2022] [Indexed: 12/31/2022] Open
Abstract
Non-human primates have an important translational value given their close phylogenetic relationship to humans. Studies in these animals remain essential for evaluating efficacy and safety of new therapeutic approaches, particularly in aging primates that display Alzheimer's disease (AD) -like pathology. With the objective to improve amyloid-β (Aβ) targeting immunotherapy, we investigated the safety and efficacy of an active immunisation with an Aβ derivative, K6Aβ1-30-NH2, in old non-human primates. Thirty-two aged (4-10 year-old) mouse lemurs were enrolled in the study, and received up to four subcutaneous injections of the vaccine in alum adjuvant or adjuvant alone. Even though antibody titres to Aβ were not high, pathological examination of the mouse lemur brains showed a significant reduction in intraneuronal Aβ that was associated with reduced microgliosis, and the vaccination did not lead to microhemorrhages. Moreover, a subtle cognitive improvement was observed in the vaccinated primates, which was probably linked to Aβ clearance. This Aβ derivative vaccine appeared to be safe as a prophylactic measure based on the brain analyses and because it did not appear to have detrimental effects on the general health of these old animals.
Collapse
Affiliation(s)
- Stéphanie G Trouche
- MMDN, Univ Montpellier, EPHE, INSERM, Montpellier, France; PSL Research University, Paris, France.
| | - Allal Boutajangout
- Departments of Neurology, and Neuroscience and Physiology, New York University Grossman School of Medicine, New York, United States.
| | - Ayodeji Asuni
- Department of Psychiatry, New York University Grossman School of Medicine, New York, United States.
| | | | - Einar M Sigurdsson
- Departments of Neuroscience and Physiology, and Psychiatry, Neuroscience Institute, New York University Grossman School of Medicine, New York, United States.
| | - Jean-Michel Verdier
- MMDN, Univ Montpellier, EPHE, INSERM, Montpellier, France; PSL Research University, Paris, France.
| | - Nadine Mestre-Francés
- MMDN, Univ Montpellier, EPHE, INSERM, Montpellier, France; PSL Research University, Paris, France.
| |
Collapse
|
3
|
Freire-Cobo C, Edler MK, Varghese M, Munger E, Laffey J, Raia S, In SS, Wicinski B, Medalla M, Perez SE, Mufson EJ, Erwin JM, Guevara EE, Sherwood CC, Luebke JI, Lacreuse A, Raghanti MA, Hof PR. Comparative neuropathology in aging primates: A perspective. Am J Primatol 2021; 83:e23299. [PMID: 34255875 PMCID: PMC8551009 DOI: 10.1002/ajp.23299] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 05/24/2021] [Accepted: 06/06/2021] [Indexed: 12/27/2022]
Abstract
While humans exhibit a significant degree of neuropathological changes associated with deficits in cognitive and memory functions during aging, non-human primates (NHP) present with more variable expressions of pathological alterations among individuals and species. As such, NHP with long life expectancy in captivity offer an opportunity to study brain senescence in the absence of the typical cellular pathology caused by age-related neurodegenerative illnesses commonly seen in humans. Age-related changes at neuronal population, single cell, and synaptic levels have been well documented in macaques and marmosets, while age-related and Alzheimer's disease-like neuropathology has been characterized in additional species including lemurs as well as great apes. We present a comparative overview of existing neuropathologic observations across the primate order, including classic age-related changes such as cell loss, amyloid deposition, amyloid angiopathy, and tau accumulation. We also review existing cellular and ultrastructural data on neuronal changes, such as dendritic attrition and spine alterations, synaptic loss and pathology, and axonal and myelin pathology, and discuss their repercussions on cellular and systems function and cognition.
Collapse
Affiliation(s)
- Carmen Freire-Cobo
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Melissa K Edler
- School of Biomedical Sciences, Kent State University, Kent, Ohio, USA
- Department of Anthropology, Kent State University, Kent, Ohio, USA
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| | - Merina Varghese
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Emily Munger
- School of Biomedical Sciences, Kent State University, Kent, Ohio, USA
- Department of Anthropology, Kent State University, Kent, Ohio, USA
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| | - Jessie Laffey
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sophia Raia
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Selena S In
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Bridget Wicinski
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Maria Medalla
- Department of Anatomy and Neurobiology, Center for Systems Neuroscience, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Sylvia E Perez
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Elliott J Mufson
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, Arizona, USA
- Department of Neurology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Joseph M Erwin
- Department of Anthropology, Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, District of Columbia, USA
| | - Elaine E Guevara
- Department of Anthropology, Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, District of Columbia, USA
- Department of Evolutionary Anthropology, Duke University, Durham, North Carolina, USA
| | - Chet C Sherwood
- Department of Anthropology, Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington, District of Columbia, USA
| | - Jennifer I Luebke
- Department of Anatomy and Neurobiology, Center for Systems Neuroscience, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Agnès Lacreuse
- Psychological and Brain Sciences, University of Massachusetts, Amherst, Massachusetts, USA
| | - Mary A Raghanti
- School of Biomedical Sciences, Kent State University, Kent, Ohio, USA
- Department of Anthropology, Kent State University, Kent, Ohio, USA
- Brain Health Research Institute, Kent State University, Kent, Ohio, USA
| | - Patrick R Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
4
|
Frye BM, Craft S, Latimer CS, Keene CD, Montine TJ, Register TC, Orr ME, Kavanagh K, Macauley SL, Shively CA. Aging-related Alzheimer's disease-like neuropathology and functional decline in captive vervet monkeys (Chlorocebus aethiops sabaeus). Am J Primatol 2021; 83:e23260. [PMID: 33818801 PMCID: PMC8626867 DOI: 10.1002/ajp.23260] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/05/2021] [Accepted: 03/21/2021] [Indexed: 12/17/2022]
Abstract
Age-related neurodegeneration characteristic of late-onset Alzheimer's disease (LOAD) begins in middle age, well before symptoms. Translational models to identify modifiable risk factors are needed to understand etiology and identify therapeutic targets. Here, we outline the evidence supporting the vervet monkey (Chlorocebus aethiops sabaeus) as a model of aging-related AD-like neuropathology and associated phenotypes including cognitive function, physical function, glucose handling, intestinal physiology, and CSF, blood, and neuroimaging biomarkers. This review provides the most comprehensive multisystem description of aging in vervets to date. This review synthesizes a large body of evidence that suggests that aging vervets exhibit a coordinated suite of traits consistent with early AD and provide a powerful, naturally occurring model for LOAD. Notably, relationships are identified between AD-like neuropathology and modifiable risk factors. Gaps in knowledge and key limitations are provided to shape future studies to illuminate mechanisms underlying divergent neurocognitive aging trajectories and to develop interventions that increase resilience to aging-associated chronic disease, particularly, LOAD.
Collapse
Affiliation(s)
- Brett M. Frye
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine
| | - Suzanne Craft
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine
- Wake Forest Alzheimer’s Disease Research Center
- J. Paul Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine
| | - Caitlin S. Latimer
- Department of Laboratory Medicine and Pathology, University of Washington-Seattle
| | - C. Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington-Seattle
| | | | - Thomas C. Register
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine
- Wake Forest Alzheimer’s Disease Research Center
- J. Paul Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine
| | - Miranda E. Orr
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine
| | - Kylie Kavanagh
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine
| | - Shannon L. Macauley
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine
- Wake Forest Alzheimer’s Disease Research Center
| | - Carol A. Shively
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine
- Wake Forest Alzheimer’s Disease Research Center
| |
Collapse
|
5
|
Kim K, Jeon HA, Seo J, Park J, Won J, Yeo HG, Jeon CY, Huh JW, Kim YH, Hong Y, Choi JW, Lee Y. Evaluation of cognitive function in adult rhesus monkeys using the finger maze test. Appl Anim Behav Sci 2020. [DOI: 10.1016/j.applanim.2020.104945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
6
|
Current status and future prospects of pathophysiology-based neuroprotective drugs for the treatment of vascular dementia. Drug Discov Today 2020; 25:793-799. [PMID: 31981482 DOI: 10.1016/j.drudis.2020.01.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 12/24/2019] [Accepted: 01/15/2020] [Indexed: 12/20/2022]
Abstract
Vascular dementia (VaD) is a progressive neurocognitive clinical syndrome that is caused by a decrease in cerebral blood flow and damage to the neurovascular unit. Given increasing life expectancy, VaD is emerging as one of the leading health problems in society. Despite the high global prevalence of cognitive impairment associated with VaD, diagnosis and treatment still remain limited because of the complexity of mechanisms of neuronal loss. Therefore, advances in our understanding of the pathophysiological mechanisms involved is crucial for the development of new therapeutic strategies. In this review, we highlight the pathophysiology, current pharmacology-based primary and secondary prevention strategies and emerging treatment options for VaD.
Collapse
|
7
|
Ruotolo R, Minato I, La Vitola P, Artioli L, Curti C, Franceschi V, Brindani N, Amidani D, Colombo L, Salmona M, Forloni G, Donofrio G, Balducci C, Del Rio D, Ottonello S. Flavonoid-Derived Human Phenyl-γ-Valerolactone Metabolites Selectively Detoxify Amyloid-β Oligomers and Prevent Memory Impairment in a Mouse Model of Alzheimer's Disease. Mol Nutr Food Res 2020; 64:e1900890. [PMID: 31914208 DOI: 10.1002/mnfr.201900890] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/24/2019] [Indexed: 11/06/2022]
Abstract
SCOPE Amyloid-β oligomers (AβO) are causally related to Alzheimer's disease (AD). Dietary natural compounds, especially flavonoids and flavan-3-ols, hold great promise as potential AD-preventive agents but their host and gut microbiota metabolism complicates identification of the most relevant bioactive species. This study aims to investigate the ability of a comprehensive set of phenyl-γ-valerolactones (PVL), the main circulating metabolites of flavan-3-ols and related dietary compounds in humans, to prevent AβO-mediated toxicity. METHODS AND RESULTS The anti-AβO activity of PVLs is examined in different cell model systems using a highly toxic β-oligomer-forming polypeptide (β23) as target toxicant. Multiple PVLs, and particularly the monohydroxylated 5-(4'-hydroxyphenyl)-γ-valerolactone metabolite [(4'-OH)-PVL], relieve β-oligomer-induced cytotoxicity in yeast and mammalian cells. As revealed by atomic force microscopy (AFM) and other in vitro assays, (4'-OH)-PVL interferes with AβO (but not fibril) assembly and actively remodels preformed AβOs into nontoxic amorphous aggregates. In keeping with the latter mode of action, treatment of AβOs with (4'-OH)-PVL prior to brain injection strongly reduces memory deterioration as well as neuroinflammation in a mouse model of AβO-induced memory impairment. CONCLUSION PVLs, which have been validated as biomarkers of the dietary intake of flavan-3-ols, lend themselves as novel AβO-selective, candidate AD-preventing compounds.
Collapse
Affiliation(s)
- Roberta Ruotolo
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124, Parma, Italy
| | - Ilaria Minato
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124, Parma, Italy
| | - Pietro La Vitola
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, 20156, Milan, Italy
| | - Luisa Artioli
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, 20156, Milan, Italy
| | - Claudio Curti
- Department of Food and Drug, University of Parma, 43124, Parma, Italy
| | | | | | - Davide Amidani
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124, Parma, Italy
| | - Laura Colombo
- Department of Molecular Biochemistry and Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, 20156, Milan, Italy
| | - Mario Salmona
- Department of Molecular Biochemistry and Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, 20156, Milan, Italy
| | - Gianluigi Forloni
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, 20156, Milan, Italy
| | - Gaetano Donofrio
- Department of Veterinary Science, University of Parma, 43126, Parma, Italy
| | - Claudia Balducci
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, 20156, Milan, Italy
| | - Daniele Del Rio
- Department of Veterinary Science, University of Parma, 43126, Parma, Italy
| | - Simone Ottonello
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124, Parma, Italy.,Biopharmanet-Tec, University of Parma, 43124, Parma, Italy
| |
Collapse
|
8
|
He Y, Ruganzu JB, Lin C, Ding B, Zheng Q, Wu X, Ma R, Liu Q, Wang Y, Jin H, Qian Y, Peng X, Ji S, Zhang L, Yang W, Lei X. Tanshinone IIA ameliorates cognitive deficits by inhibiting endoplasmic reticulum stress-induced apoptosis in APP/PS1 transgenic mice. Neurochem Int 2019; 133:104610. [PMID: 31778727 DOI: 10.1016/j.neuint.2019.104610] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/15/2019] [Accepted: 11/23/2019] [Indexed: 02/06/2023]
Abstract
Our previous data indicated that tanshinone IIA (tan IIA) improves learning and memory in a mouse model of Alzheimer's disease (AD) induced by streptozotocin via restoring cholinergic function, attenuating oxidative stress and blocking p38 MAPK signal pathway activation. This study aims to estimate whether tan IIA inhibits endoplasmic reticulum (ER) stress-induced apoptosis to prevent cognitive decline in APP/PS1 transgenic mice. Tan IIA (10 mg/kg and 30 mg/kg) was intraperitoneally administered to the six-month-old APP/PS1 mice for 30 consecutive days. β-amyloid (Aβ) plaques were measured by immunohistochemisty and Thioflavin S staining, apoptotic cells were observed by TUNEL, ER stress markers and apoptosis signaling proteins were investigated by western blotting and RT-PCR. Our results showed that tan IIA significantly ameliorates cognitive deficits and improves spatial learning ability of APP/PS1 mice in the nest-building test, novel object recognition test and Morris water maze test. Furthermore, tan IIA significantly reduced the deposition of Aβ plaques and neuronal apoptosis, and markedly prevented abnormal expression of glucose regulated protein 78 (GRP78), initiation factor 2α (eIF2α), inositol-requiring enzyme 1α (IRE1α), activating transcription factor 6 (ATF6), as well as suppressed the activation of C/EBP homologous protein (CHOP) and c-Jun N-terminal kinase (JNK) pathways in the parietal cortex and hippocampus. Moreover, tan IIA induced an up-regulation of the Bcl-2/Bax ratio and down-regulation of caspase-3 protein activity. Taken together, the above findings indicated that tan IIA improves learning and memory through attenuating Aβ plaques deposition and inhibiting ER stress-induced apoptosis. These results suggested that tan IIA might become a promising therapeutic candidate drug against AD.
Collapse
Affiliation(s)
- Yingying He
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - John Bosco Ruganzu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Chengheng Lin
- Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Bo Ding
- Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Quzhao Zheng
- Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Xiangyuan Wu
- Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Ruiyang Ma
- Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Qian Liu
- Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Yang Wang
- Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Hui Jin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Yihua Qian
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Xiaoqian Peng
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Shengfeng Ji
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Liangliang Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi province, 710061, China
| | - Weina Yang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China.
| | - Xiaomei Lei
- Department of Child Health Care, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi province, 710004, China.
| |
Collapse
|
9
|
Drummond E, Goñi F, Liu S, Prelli F, Scholtzova H, Wisniewski T. Potential Novel Approaches to Understand the Pathogenesis and Treat Alzheimer's Disease. J Alzheimers Dis 2019; 64:S299-S312. [PMID: 29562516 DOI: 10.3233/jad-179909] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There is growing genetic and proteomic data highlighting the complexity of Alzheimer's disease (AD) pathogenesis. Greater use of unbiased "omics" approaches is being increasingly recognized as essential for the future development of effective AD research, that need to better reflect the multiple distinct pathway abnormalities that can drive AD pathology. The track record of success in AD clinical trials thus far has been very poor. In part, this high failure rate has been related to the premature translation of highly successful results in animal models that mirror only limited aspects of AD pathology to humans. We highlight our recent efforts to increase use of human tissue to gain a better understanding of the AD pathogenesis subtype variety and to develop several distinct therapeutic approaches tailored to address this diversity. These therapeutic approaches include the blocking of the Aβ/apoE interaction, stimulation of innate immunity, and the simultaneous blocking of Aβ/tau oligomer toxicity. We believe that future successful therapeutic approaches will need to be combined to better reflect the complexity of the abnormal pathways triggered in AD pathogenesis.
Collapse
Affiliation(s)
- Eleanor Drummond
- Department of Neurology, Center for Cognitive Neurology, NYU School of Medicine, New York, NY, USA
| | - Fernando Goñi
- Department of Neurology, Center for Cognitive Neurology, NYU School of Medicine, New York, NY, USA
| | - Shan Liu
- Department of Neurology, Center for Cognitive Neurology, NYU School of Medicine, New York, NY, USA
| | - Frances Prelli
- Department of Neurology, Center for Cognitive Neurology, NYU School of Medicine, New York, NY, USA
| | - Henrieta Scholtzova
- Department of Neurology, Center for Cognitive Neurology, NYU School of Medicine, New York, NY, USA
| | - Thomas Wisniewski
- Departments of Neurology, Pathology and Psychiatry, Center for Cognitive Neurology, NYU School of Medicine, New York, NY, USA
| |
Collapse
|
10
|
Surguchov A, Emamzadeh FN, Surguchev AA. Amyloidosis and Longevity: A Lesson from Plants. BIOLOGY 2019; 8:biology8020043. [PMID: 31137746 PMCID: PMC6628237 DOI: 10.3390/biology8020043] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/16/2019] [Accepted: 05/22/2019] [Indexed: 12/16/2022]
Abstract
The variety of lifespans of different organisms in nature is amazing. Although it is acknowledged that the longevity is determined by a complex interaction between hereditary and environmental factors, many questions about factors defining lifespan remain open. One of them concerns a wide range of lifespans of different organisms. The reason for the longevity of certain trees, which reaches a thousand years and exceeds the lifespan of most long living vertebrates by a huge margin is also not completely understood. Here we have discussed some distinguishing characteristics of plants, which may explain their remarkable longevity. Among them are the absence (or very low abundance) of intracellular inclusions composed of amyloidogenic proteins, the lack of certain groups of proteins prone to aggregate and form amyloids in animals, and the high level of compounds which inhibit protein aggregation and possess antiaging properties.
Collapse
Affiliation(s)
- Andrei Surguchov
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - Fatemeh Nouri Emamzadeh
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, University of Lancaster, Lancaster LA1 4AY, UK.
| | - Alexei A Surguchev
- Section of Otolaryngology, Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
11
|
Panza F, Lozupone M, Logroscino G, Imbimbo BP. A critical appraisal of amyloid-β-targeting therapies for Alzheimer disease. Nat Rev Neurol 2019; 15:73-88. [DOI: 10.1038/s41582-018-0116-6] [Citation(s) in RCA: 459] [Impact Index Per Article: 91.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
12
|
Wisniewski T, Drummond E. Future horizons in Alzheimer's disease research. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 168:223-241. [PMID: 31699317 DOI: 10.1016/bs.pmbts.2019.08.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
There are growing genetic, transcriptomic and proteomic data pointing to the complexity of Alzheimer's disease (AD) pathogenesis. Unbiased "omics" approaches are essential for the future development of effective AD research, which will need to be combined and personalized, given that multiple distinct pathways can drive AD pathology. It is essential to gain a better understanding of the AD pathogenesis subtype variety and to develop several distinct therapeutic approaches tailored to address this diversity, as well as the common presence of mixed pathologies. These nonmutually exclusive therapeutic approaches include the targeting of multiple toxic oligomeric species concurrently, targeting the apolipoprotein E/amyloid β interaction and the modulation of innate immunity, as well as more "out of the box" ideas such as targeting infectious agents that may play a role in AD.
Collapse
Affiliation(s)
- Thomas Wisniewski
- Departments of Neurology, Pathology and Psychiatry, Center for Cognitive Neurology, NYU School of Medicine, New York, NY, United States.
| | - Eleanor Drummond
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
13
|
Kozlowski PA, Aldovini A. Mucosal Vaccine Approaches for Prevention of HIV and SIV Transmission. CURRENT IMMUNOLOGY REVIEWS 2019; 15:102-122. [PMID: 31452652 PMCID: PMC6709706 DOI: 10.2174/1573395514666180605092054] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 04/19/2018] [Accepted: 05/30/2018] [Indexed: 02/06/2023]
Abstract
Optimal protective immunity to HIV will likely require that plasma cells, memory B cells and memory T cells be stationed in mucosal tissues at portals of viral entry. Mucosal vaccine administration is more effective than parenteral vaccine delivery for this purpose. The challenge has been to achieve efficient vaccine uptake at mucosal surfaces, and to identify safe and effective adjuvants, especially for mucosally administered HIV envelope protein immunogens. Here, we discuss strategies used to deliver potential HIV vaccine candidates in the intestine, respiratory tract, and male and female genital tract of humans and nonhuman primates. We also review mucosal adjuvants, including Toll-like receptor agonists, which may adjuvant both mucosal humoral and cellular immune responses to HIV protein immunogens.
Collapse
Affiliation(s)
- Pamela A. Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Anna Aldovini
- Department of Medicine, and Harvard Medical School, Boston Children’s Hospital, Department of Pediatrics, Boston MA, 02115, USA
| |
Collapse
|
14
|
Pu Z, Ma S, Wang L, Li M, Shang L, Luo Y, Chen W. Amyloid-beta Degradation and Neuroprotection of Dauricine Mediated by Unfolded Protein Response in a Caenorhabditis elegans Model of Alzheimer’s disease. Neuroscience 2018; 392:25-37. [DOI: 10.1016/j.neuroscience.2018.09.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 09/06/2018] [Accepted: 09/17/2018] [Indexed: 01/04/2023]
|
15
|
Xu J, Wang K, Yuan Y, Li H, Zhang R, Guan S, Wang L. A Novel Peroxidase Mimics and Ameliorates Alzheimer's Disease-Related Pathology and Cognitive Decline in Mice. Int J Mol Sci 2018; 19:ijms19113304. [PMID: 30352982 PMCID: PMC6274722 DOI: 10.3390/ijms19113304] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 10/14/2018] [Accepted: 10/19/2018] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder in the elderly, which is characterized by the accumulation of amyloid β (Aβ) plaques, oxidative stress, and neuronal loss. Therefore, clearing Aβ aggregates and reducing oxidative stress could be an effective therapeutic strategy for AD. Deuterohemin-AlaHisThrValGluLys (DhHP-6), a novel deuterohemin-containing peptide mimetic of the natural microperoxidase-11 (MP-11), shows higher antioxidant activity and stability compared to the natural microperoxidases. DhHP-6 possesses the ability of extending lifespan and alleviating paralysis in the Aβ1-42 transgenic Caenorhabditis elegans CL4176 model of AD, as shown in our previous study. Therefore, this study was aimed at exploring the neuroprotective effect of DhHP-6 in the APPswe/PSEN1dE9 transgenic mouse model of AD. DhHP-6 reduced the diameter and fiber structure of Aβ1-42 aggregation in vitro, as shown by dynamic light scattering and transmission electron microscope. DhHP-6 exerted its neuroprotective effect by inhibiting Aβ aggregation and plaque formation, and by reducing Aβ1-42 oligomers-induced neurotoxicity on HT22 (mouse hippocampal neuronal) and SH-SY5Y (human neuroblastoma) cells. In the AD mouse model, DhHP-6 significantly ameliorated cognitive decline and improved spatial learning ability in behavioral tests including the Morris water maze, Y-maze, novel object recognition, open field, and nest-building test. Moreover, DhHP-6 reduced the deposition of Aβ plaques in the cerebral cortex and hippocampus. More importantly, DhHP-6 restored the morphology of astrocytes and microglia, and significantly reduced the levels of pro-inflammatory cytokines. Our findings provide a basis for considering the non-toxic, peroxidase mimetic DhHP-6 as a new candidate drug against AD.
Collapse
Affiliation(s)
- Jia Xu
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Kai Wang
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Ye Yuan
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Hui Li
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Ruining Zhang
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Shuwen Guan
- School of Life Sciences, Jilin University, Changchun 130012, China.
- Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun 130012, China.
- Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, Jilin Universtiy, Changchun 130012, China.
| | - Liping Wang
- School of Life Sciences, Jilin University, Changchun 130012, China.
- Engineering Laboratory for AIDS Vaccine, Jilin University, Changchun 130012, China.
- Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, Jilin Universtiy, Changchun 130012, China.
| |
Collapse
|
16
|
Martins YA, Tsuchida CJ, Antoniassi P, Demarchi IG. Efficacy and Safety of the Immunization with DNA for Alzheimer's Disease in Animal Models: A Systematic Review from Literature. J Alzheimers Dis Rep 2017; 1:195-217. [PMID: 30480238 PMCID: PMC6159633 DOI: 10.3233/adr-170025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disease that does not have a proven cure; however, one of the most promising strategies for its treatment has been DNA vaccines. OBJECTIVE The present review is aimed to report the new developments of the efficacy and safety of DNA vaccines for AD in animal models. METHOD The method PRISMA was used for this review. The article search was made in the electronic databases PubMed, LILACS, and Scopus using the descriptors ''Alzheimer disease" and ''Vaccine, DNA". Articles published between January 2001 and September 2017 in English, Portuguese, and Spanish were included. RESULTS Upon the consensus, the researchers identified 28 original articles. The studies showed satisfying results as for the decrease of amyloid plaques in mouse, rabbits, and monkeys brains using mostly the DNA Aβ42 vaccine, AV-1955, and AdPEDI-(Aβ1-6)11, mainly with a gene gun. In addition to a reduction in tau by the first DNA vaccine (AV-1980D) targeting this protein. The use of adjuvants and boosters also had positive results as they increased the destruction of the amyloid plaques and induced an anti-inflammatory response profile without side effects. CONCLUSION The results of DNA vaccines targeting the amyloid-β and the tau protein with or without adjuvants and boosters were promising in reducing amyloid plaques and tau protein without side effects in animals. Although there are many vaccines being tested in animals, few reach clinical trials. Thus, as a future perspective, we suggest that clinical studies should be conducted with vaccines that have been promising in animal models (e.g., DNA Aβ42 vaccine, AV-1955, and AdPEDI-(Aβ1-6)11).
Collapse
|
17
|
Panza F, Seripa D, Lozupone M, Solfrizzi V, Imbimbo BP, Barulli MR, Tortelli R, Capozzo R, Bisceglia P, Dimitri A, Stallone R, Dibello V, Quaranta N, Daniele A, Bellomo A, Greco A, Logroscino G. The potential of solanezumab and gantenerumab to prevent Alzheimer’s disease in people with inherited mutations that cause its early onset. Expert Opin Biol Ther 2017; 18:25-35. [DOI: 10.1080/14712598.2018.1389885] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Francesco Panza
- Unit of Neurodegenerative Disease, Department of Basic Medicine Sciences, Neuroscience, and Sense Organs, University of Bari ‘Aldo Moro’, Bari, Italy
- Unit of Neurodegenerative Disease, Department of Clinical Research in Neurology, University of Bari ‘Aldo Moro’ at ‘Pia Fondazione Card. G. Panico’, Tricase, Italy
- Geriatric Unit and Gerontology-Geriatrics Research Laboratory, Department of Medical Sciences, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Davide Seripa
- Geriatric Unit and Gerontology-Geriatrics Research Laboratory, Department of Medical Sciences, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Madia Lozupone
- Unit of Neurodegenerative Disease, Department of Basic Medicine Sciences, Neuroscience, and Sense Organs, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Vincenzo Solfrizzi
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Bruno P. Imbimbo
- Research & Development Department, Chiesi Farmaceutici, Parma, Italy
| | - Maria Rosaria Barulli
- Unit of Neurodegenerative Disease, Department of Clinical Research in Neurology, University of Bari ‘Aldo Moro’ at ‘Pia Fondazione Card. G. Panico’, Tricase, Italy
| | - Rosanna Tortelli
- Unit of Neurodegenerative Disease, Department of Clinical Research in Neurology, University of Bari ‘Aldo Moro’ at ‘Pia Fondazione Card. G. Panico’, Tricase, Italy
| | - Rosa Capozzo
- Unit of Neurodegenerative Disease, Department of Clinical Research in Neurology, University of Bari ‘Aldo Moro’ at ‘Pia Fondazione Card. G. Panico’, Tricase, Italy
| | - Paola Bisceglia
- Geriatric Unit and Gerontology-Geriatrics Research Laboratory, Department of Medical Sciences, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Andrea Dimitri
- Psychiatric Unit, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Roberta Stallone
- Unit of Neurodegenerative Disease, Department of Basic Medicine Sciences, Neuroscience, and Sense Organs, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Vittorio Dibello
- Interdisciplinary Department of Medicine (DIM), Section of Dentistry, University of Bari Aldo, Moro, Italy
| | - Nicola Quaranta
- Otolaryngology Unit, University of Bari Aldo Moro, Bari, Italy
| | - Antonio Daniele
- Institute of Neurology, Catholic University of Sacred Heart, Rome, Italy
| | - Antonello Bellomo
- Psychiatric Unit, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Antonio Greco
- Geriatric Unit and Gerontology-Geriatrics Research Laboratory, Department of Medical Sciences, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Giancarlo Logroscino
- Unit of Neurodegenerative Disease, Department of Basic Medicine Sciences, Neuroscience, and Sense Organs, University of Bari ‘Aldo Moro’, Bari, Italy
- Unit of Neurodegenerative Disease, Department of Clinical Research in Neurology, University of Bari ‘Aldo Moro’ at ‘Pia Fondazione Card. G. Panico’, Tricase, Italy
| |
Collapse
|
18
|
Production of Monoclonal Antibodies to Pathologic β-sheet Oligomeric Conformers in Neurodegenerative Diseases. Sci Rep 2017; 7:9881. [PMID: 28852189 PMCID: PMC5575137 DOI: 10.1038/s41598-017-10393-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 08/07/2017] [Indexed: 01/12/2023] Open
Abstract
We describe a novel approach to produce conformational monoclonal antibodies selected to specifically react with the β-sheet secondary structure of pathological oligomeric conformers, characteristic of many neurodegenerative diseases. Contrary to past and current efforts, we utilize a mammalian non-self-antigen as an immunogen. The small, non-self peptide selected was covalently polymerized with glutaraldehyde until it reached a high β-sheet secondary structure content, and species between 10–100kDa that are immunogenic, stable and soluble (p13Bri). Inoculation of p13Bri in mice elicited antibodies to the peptide and the β-sheet secondary structure conformation. Hybridomas were produced and clones selected for their reactivity with at least two different oligomeric conformers from Alzheimer’s, Parkinson and/or Prion diseases. The resulting conformational monoclonals are able to detect pathological oligomeric forms in different human neurodegenerative diseases by ELISA, immunohistochemistry and immunoblots. This technological approach may be useful to develop tools for detection, monitoring and treatment of multiple misfolding disorders.
Collapse
|
19
|
Liu S, Park S, Allington G, Prelli F, Sun Y, Martá-Ariza M, Scholtzova H, Biswas G, Brown B, Verghese PB, Mehta PD, Kwon YU, Wisniewski T. Targeting Apolipoprotein E/Amyloid β Binding by Peptoid CPO_Aβ17-21 P Ameliorates Alzheimer's Disease Related Pathology and Cognitive Decline. Sci Rep 2017; 7:8009. [PMID: 28808293 PMCID: PMC5556019 DOI: 10.1038/s41598-017-08604-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 07/12/2017] [Indexed: 02/01/2023] Open
Abstract
Inheritance of the apolipoprotein E4 (apoE4) genotype has been identified as the major genetic risk factor for late onset Alzheimer's disease (AD). Studies have shown that apoE, apoE4 in particular, binds to amyloid-β (Aβ) peptides at residues 12-28 of Aβ and this binding modulates Aβ accumulation and disease progression. We have previously shown in several AD transgenic mice lines that blocking the apoE/Aβ interaction with Aβ12-28 P reduced Aβ and tau-related pathology, leading to cognitive improvements in treated AD mice. Recently, we have designed a small peptoid library derived from the Aβ12-28 P sequence to screen for new apoE/Aβ binding inhibitors with higher efficacy and safety. Peptoids are better drug candidates than peptides due to their inherently more favorable pharmacokinetic properties. One of the lead peptoid compounds, CPO_Aβ17-21 P, diminished the apoE/Aβ interaction and attenuated the apoE4 pro-fibrillogenic effects on Aβ aggregation in vitro as well as apoE4 potentiation of Aβ cytotoxicity. CPO_Aβ17-21 P reduced Aβ-related pathology coupled with cognitive improvements in an AD APP/PS1 transgenic mouse model. Our study suggests the non-toxic, non-fibrillogenic peptoid CPO_Aβ17-21 P has significant promise as a new AD therapeutic agent which targets the Aβ related apoE pathway, with improved efficacy and pharmacokinetic properties.
Collapse
Affiliation(s)
- Shan Liu
- Center for Cognitive Neurology, Department of Neurology, New York University School of Medicine, New York, USA
| | - Shinae Park
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, Korea
| | | | - Frances Prelli
- Center for Cognitive Neurology, Department of Neurology, New York University School of Medicine, New York, USA
| | - Yanjie Sun
- Center for Cognitive Neurology, Department of Neurology, New York University School of Medicine, New York, USA
| | - Mitchell Martá-Ariza
- Center for Cognitive Neurology, Department of Neurology, New York University School of Medicine, New York, USA
| | - Henrieta Scholtzova
- Center for Cognitive Neurology, Department of Neurology, New York University School of Medicine, New York, USA
| | - Goutam Biswas
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, Korea
| | - Bernard Brown
- Center for Cognitive Neurology, Department of Neurology, New York University School of Medicine, New York, USA
| | - Philip B Verghese
- C2N Diagnostics, Center for Emerging Technologies, 4041 Forest Park Avenue, St. Louis, MO, 63108, USA
| | - Pankaj D Mehta
- Department of Immunology, New York State Institute for Basic Research in Developmental Disabilities, New York, USA
| | - Yong-Uk Kwon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, Korea.
| | - Thomas Wisniewski
- Center for Cognitive Neurology, Departments of Neurology, Psychiatry and Pathology, Neuroscience Institute, New York University School of Medicine, New York, USA.
| |
Collapse
|