1
|
Moon HJ, Luo Y, Chugh D, Zhao L. Human apolipoprotein E glycosylation and sialylation: from structure to function. Front Mol Neurosci 2024; 17:1399965. [PMID: 39169951 PMCID: PMC11335735 DOI: 10.3389/fnmol.2024.1399965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/28/2024] [Indexed: 08/23/2024] Open
Abstract
Human apolipoprotein E (ApoE) was first identified as a polymorphic gene in the 1970s; however, the genetic association of ApoE genotypes with late-onset sporadic Alzheimer's disease (sAD) was only discovered 20 years later. Since then, intensive research has been undertaken to understand the molecular effects of ApoE in the development of sAD. Despite three decades' worth of effort and over 10,000 papers published, the greatest mystery in the ApoE field remains: human ApoE isoforms differ by only one or two amino acid residues; what is responsible for their significantly distinct roles in the etiology of sAD, with ApoE4 conferring the greatest genetic risk for sAD whereas ApoE2 providing exceptional neuroprotection against sAD. Emerging research starts to point to a novel and compelling hypothesis that the sialoglycans posttranslationally appended to human ApoE may serve as a critical structural modifier that alters the biology of ApoE, leading to the opposing impacts of ApoE isoforms on sAD and likely in the peripheral systems as well. ApoE has been shown to be posttranslationally glycosylated in a species-, tissue-, and cell-specific manner. Human ApoE, particularly in brain tissue and cerebrospinal fluid (CSF), is highly glycosylated, and the glycan chains are exclusively attached via an O-linkage to serine or threonine residues. Moreover, studies have indicated that human ApoE glycans undergo sialic acid modification or sialylation, a structural alteration found to be more prominent in ApoE derived from the brain and CSF than plasma. However, whether the sialylation modification of human ApoE has a biological role is largely unexplored. Our group recently first reported that the three major isoforms of human ApoE in the brain undergo varying degrees of sialylation, with ApoE2 exhibiting the most abundant sialic acid modification, whereas ApoE4 is the least sialylated. Our findings further indicate that the sialic acid moiety on human ApoE glycans may serve as a critical modulator of the interaction of ApoE with amyloid β (Aβ) and downstream Aβ pathogenesis, a prominent pathologic feature in AD. In this review, we seek to provide a comprehensive summary of this exciting and rapidly evolving area of ApoE research, including the current state of knowledge and opportunities for future exploration.
Collapse
Affiliation(s)
- Hee-Jung Moon
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Yan Luo
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Diksha Chugh
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Liqin Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
- Neuroscience Graduate Program, University of Kansas, Lawrence, KS, United States
| |
Collapse
|
2
|
Zhang H, Gomika Udugamasooriya D. Linker optimization and activity validation of a cell surface vimentin targeted homo-dimeric peptoid antagonist for lung cancer stem cells. Bioorg Med Chem 2024; 97:117560. [PMID: 38103535 DOI: 10.1016/j.bmc.2023.117560] [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: 10/24/2023] [Revised: 12/01/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT) endows epithelia-derived cancer cells with properties of stem cells that govern cancer invasion and metastasis. Vimentin is one of the best studied EMT markers and recent reports indicate that vimentin interestingly translocated onto cell surface under various tumor conditions. We recently reported a cell surface vimentin (CSV) specific peptoid antagonist named JM3A. We now investigated the selective antagonist activity of the optimized homo-dimeric version of JM3A, JM3A-L2D on stem-like cancer cells or cancer stem cells (CSCs) over normal cells in non-small cell lung cancer (NSCLC). Homo-dimerization of JM3A provided the avidity effect and improved the biological activity compared to the monomeric version. We first optimized the central linker length of the dimer by designing seven JM3A derivatives with varying linker lengths/types and evaluated the anti-cancer activity using the standard MTS cell viability assay. The most optimized derivative contains a central lysine linker and two glycines, named JM3A-L2D, which displayed 100 nM vimentin binding affinity (Kd) with an anti-cancer activity (IC50) of 6.7 µM on H1299 NSCLC cells. This is a 190-fold improvement in binding over the original JM3A. JM3A-L2D exhibited better potency on high vimentin-expressing NSCLC cells (H1299 and H460) compared to low vimentin-expressing NSCLC cells (H2122). No activity was observed on normal bronchial HBEC3-KT cells. The anti-CSC activity of JM3A-L2D was evaluated using the standard colony formation assay and JM3A-L2D disrupted the colony formation with IC50 ∼ 400 nM. In addition, JM3A-L2D inhibited cell migration activity at IC50 ∼ 2 µM, assessed via wound healing assay. The underlying mechanism of action seems to be the induction of apoptosis by JM3A-L2D on high-vimentin expressing H1229 and H460 NSCLC cells. Our optimized highly CSV selective peptoid has the potential to be developed as an anti-cancer drug candidate, especially considering the high serum stability and economical synthesis of peptoids.
Collapse
Affiliation(s)
- Haowen Zhang
- Department of Pharmacological & Pharmaceutical Sciences, University of Houston, 4349 Martin Luther King Boulevard, Health Building 2, Room 7033, Houston, TX 77204-5037, USA
| | - D Gomika Udugamasooriya
- Department of Pharmacological & Pharmaceutical Sciences, University of Houston, 4349 Martin Luther King Boulevard, Health Building 2, Room 7033, Houston, TX 77204-5037, USA; Department of Cancer Systems Imaging, MD Anderson Cancer Center, 1881 East Road, Houston, TX 77030-4009, USA.
| |
Collapse
|
3
|
Perluigi M, Di Domenico F, Butterfield DA. Oxidative damage in neurodegeneration: roles in the pathogenesis and progression of Alzheimer disease. Physiol Rev 2024; 104:103-197. [PMID: 37843394 PMCID: PMC11281823 DOI: 10.1152/physrev.00030.2022] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 03/30/2023] [Accepted: 05/24/2023] [Indexed: 10/17/2023] Open
Abstract
Alzheimer disease (AD) is associated with multiple etiologies and pathological mechanisms, among which oxidative stress (OS) appears as a major determinant. Intriguingly, OS arises in various pathways regulating brain functions, and it seems to link different hypotheses and mechanisms of AD neuropathology with high fidelity. The brain is particularly vulnerable to oxidative damage, mainly because of its unique lipid composition, resulting in an amplified cascade of redox reactions that target several cellular components/functions ultimately leading to neurodegeneration. The present review highlights the "OS hypothesis of AD," including amyloid beta-peptide-associated mechanisms, the role of lipid and protein oxidation unraveled by redox proteomics, and the antioxidant strategies that have been investigated to modulate the progression of AD. Collected studies from our groups and others have contributed to unraveling the close relationships between perturbation of redox homeostasis in the brain and AD neuropathology by elucidating redox-regulated events potentially involved in both the pathogenesis and progression of AD. However, the complexity of AD pathological mechanisms requires an in-depth understanding of several major intracellular pathways affecting redox homeostasis and relevant for brain functions. This understanding is crucial to developing pharmacological strategies targeting OS-mediated toxicity that may potentially contribute to slow AD progression as well as improve the quality of life of persons with this severe dementing disorder.
Collapse
Affiliation(s)
- Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
4
|
Canepa E, Parodi-Rullan R, Vazquez-Torres R, Gamallo-Lana B, Guzman-Hernandez R, Lemon NL, Angiulli F, Debure L, Ilies MA, Østergaard L, Wisniewski T, Gutiérrez-Jiménez E, Mar AC, Fossati S. FDA-approved carbonic anhydrase inhibitors reduce amyloid β pathology and improve cognition, by ameliorating cerebrovascular health and glial fitness. Alzheimers Dement 2023; 19:5048-5073. [PMID: 37186121 PMCID: PMC10600328 DOI: 10.1002/alz.13063] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 05/17/2023]
Abstract
INTRODUCTION Cerebrovascular pathology is an early and causal hallmark of Alzheimer's disease (AD), in need of effective therapies. METHODS Based on the success of our previous in vitro studies, we tested for the first time in a model of AD and cerebral amyloid angiopathy (CAA), the carbonic anhydrase inhibitors (CAIs) methazolamide and acetazolamide, Food and Drug Administration-approved against glaucoma and high-altitude sickness. RESULTS Both CAIs reduced cerebral, vascular, and glial amyloid beta (Aβ) accumulation and caspase activation, diminished gliosis, and ameliorated cognition in TgSwDI mice. The CAIs also improved microvascular fitness and induced protective glial pro-clearance pathways, resulting in the reduction of Aβ deposition. Notably, we unveiled that the mitochondrial carbonic anhydrase-VB (CA-VB) is upregulated in TgSwDI brains, CAA and AD+CAA human subjects, and in endothelial cells upon Aβ treatment. Strikingly, CA-VB silencing specifically reduces Aβ-mediated endothelial apoptosis. DISCUSSION This work substantiates the potential application of CAIs in clinical trials for AD and CAA.
Collapse
Affiliation(s)
- Elisa Canepa
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Rebecca Parodi-Rullan
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Rafael Vazquez-Torres
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Begona Gamallo-Lana
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Roberto Guzman-Hernandez
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Nicole L. Lemon
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Federica Angiulli
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Ludovic Debure
- Department on Neurology, Center for Cognitive Neurology, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Marc A. Ilies
- Department of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, Temple University, Philadelphia, PA, 19140, USA
| | - Leif Østergaard
- Center of Functionally Integrative Neuroscience (CFIN), Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Thomas Wisniewski
- Department on Neurology, Center for Cognitive Neurology, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Eugenio Gutiérrez-Jiménez
- Center of Functionally Integrative Neuroscience (CFIN), Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Adam C. Mar
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Silvia Fossati
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| |
Collapse
|
5
|
Puranik N, Yadav D, Song M. Advancements in the Application of Nanomedicine in Alzheimer's Disease: A Therapeutic Perspective. Int J Mol Sci 2023; 24:14044. [PMID: 37762346 PMCID: PMC10530821 DOI: 10.3390/ijms241814044] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/11/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that affects most people worldwide. AD is a complex central nervous system disorder. Several drugs have been designed to cure AD, but with low success rates. Because the blood-brain and blood-cerebrospinal fluid barriers are two barriers that protect the central nervous system, their presence has severely restricted the efficacy of many treatments that have been studied for AD diagnosis and/or therapy. The use of nanoparticles for the diagnosis and treatment of AD is the focus of an established and rapidly developing field of nanomedicine. Recent developments in nanomedicine have made it possible to effectively transport drugs to the brain. However, numerous obstacles remain to the successful use of nanomedicines in clinical settings for AD treatment. Furthermore, given the rapid advancement in nanomedicine therapeutics, better outcomes for patients with AD can be anticipated. This article provides an overview of recent developments in nanomedicine using different types of nanoparticles for the management and treatment of AD.
Collapse
Affiliation(s)
| | | | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (N.P.); (D.Y.)
| |
Collapse
|
6
|
Arar S, Haque MA, Kayed R. Protein aggregation and neurodegenerative disease: Structural outlook for the novel therapeutics. Proteins 2023:10.1002/prot.26561. [PMID: 37530227 PMCID: PMC10834863 DOI: 10.1002/prot.26561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 08/03/2023]
Abstract
Before the controversial approval of humanized monoclonal antibody lecanemab, which binds to the soluble amyloid-β protofibrils, all the treatments available earlier, for Alzheimer's disease (AD) were symptomatic. The researchers are still struggling to find a breakthrough in AD therapeutic medicine, which is partially attributable to lack in understanding of the structural information associated with the intrinsically disordered proteins and amyloids. One of the major challenges in this area of research is to understand the structural diversity of intrinsically disordered proteins under in vitro conditions. Therefore, in this review, we have summarized the in vitro applications of biophysical methods, which are aimed to shed some light on the heterogeneity, pathogenicity, structures and mechanisms of the intrinsically disordered protein aggregates associated with proteinopathies including AD. This review will also rationalize some of the strategies in modulating disease-relevant pathogenic protein entities by small molecules using structural biology approaches and biophysical characterization. We have also highlighted tools and techniques to simulate the in vivo conditions for native and cytotoxic tau/amyloids assemblies, urge new chemical approaches to replicate tau/amyloids assemblies similar to those in vivo conditions, in addition to designing novel potential drugs.
Collapse
Affiliation(s)
- Sharif Arar
- Mitchell Center for Neurodegenerative Diseases
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, 77555, USA
- Department of Chemistry, School of Science, The University of Jordan, Amman 11942, Jordan
| | - Md Anzarul Haque
- Mitchell Center for Neurodegenerative Diseases
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| |
Collapse
|
7
|
Valles SL, Singh SK, Campos-Campos J, Colmena C, Campo-Palacio I, Alvarez-Gamez K, Caballero O, Jorda A. Functions of Astrocytes under Normal Conditions and after a Brain Disease. Int J Mol Sci 2023; 24:ijms24098434. [PMID: 37176144 PMCID: PMC10179527 DOI: 10.3390/ijms24098434] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/26/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
In the central nervous system (CNS) there are a greater number of glial cells than neurons (between five and ten times more). Furthermore, they have a greater number of functions (more than eight functions). Glia comprises different types of cells, those of neural origin (astrocytes, radial glia, and oligodendroglia) and differentiated blood monocytes (microglia). During ontogeny, neurons develop earlier (at fetal day 15 in the rat) and astrocytes develop later (at fetal day 21 in the rat), which could indicate their important and crucial role in the CNS. Analysis of the phylogeny reveals that reptiles have a lower number of astrocytes compared to neurons and in humans this is reversed, as there have a greater number of astrocytes compared to neurons. These data perhaps imply that astrocytes are important and special cells, involved in many vital functions, including memory, and learning processes. In addition, astrocytes are involved in different mechanisms that protect the CNS through the production of antioxidant and anti-inflammatory proteins and they clean the extracellular environment and help neurons to communicate correctly with each other. The production of inflammatory mediators is important to prevent changes in brain homeostasis. On the contrary, excessive, or continued production appears as a characteristic element in many diseases, such as Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), and in neurodevelopmental diseases, such as bipolar disorder, schizophrenia, and autism. Furthermore, different drugs and techniques have been developed to reverse oxidative stress and/or excess of inflammation that occurs in many CNS diseases, but much remains to be investigated. This review attempts to highlight the functional relevance of astrocytes in normal and neuropathological conditions by showing the molecular and cellular mechanisms of their role in the CNS.
Collapse
Affiliation(s)
- Soraya L Valles
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
| | - Sandeep Kumar Singh
- Indian Scientific Education and Technology Foundation, Lucknow 226002, India
| | - Juan Campos-Campos
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
- Faculty of Nursing and Podiatry, University of Valencia, 46010 Valencia, Spain
| | - Carlos Colmena
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
| | - Ignacio Campo-Palacio
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
| | - Kenia Alvarez-Gamez
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
| | - Oscar Caballero
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
- Faculty of Nursing and Podiatry, University of Valencia, 46010 Valencia, Spain
| | - Adrian Jorda
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
- Faculty of Nursing and Podiatry, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
8
|
Qiu W, Liu H, Liu Y, Lu X, Wang L, Hu Y, Feng F, Li Q, Sun H. Regulation of beta-amyloid for the treatment of Alzheimer's disease: Research progress of therapeutic strategies and bioactive compounds. Med Res Rev 2023. [PMID: 36945751 DOI: 10.1002/med.21947] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/09/2023] [Accepted: 02/26/2023] [Indexed: 03/23/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that is difficult to treat. Extracellular amyloid is the principal pathological criterion for the diagnosis of AD. Amyloid β (Aβ) interacts with various receptor molecules on the plasma membrane and mediates a series of signaling pathways that play a vital role in the occurrence and development of AD. Research on receptors that interact with Aβ is currently ongoing. Overall, there are no effective medications to treat AD. In this review, we first discuss the importance of Aβ in the pathogenesis of AD, then summarize the latest progress of Aβ-related targets and compounds. Finally, we put forward the challenges and opportunities in the development of effective AD therapies.
Collapse
Affiliation(s)
- Weimin Qiu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hui Liu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yijun Liu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xin Lu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Lei Wang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yanyu Hu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
- Department of Natural Medicinal Chemistry, Jiangsu Food and Pharmaceuticals Science College, Institute of Food and Pharmaceuticals Research, Jiangsu, Huaian, China
| | - Qi Li
- Department of Pharmacology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Haopeng Sun
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
9
|
Lazarev VF, Dutysheva EA, Kanunikov IE, Guzhova IV, Margulis BA. Protein Interactome of Amyloid-β as a Therapeutic Target. Pharmaceuticals (Basel) 2023; 16:312. [PMID: 37259455 PMCID: PMC9965366 DOI: 10.3390/ph16020312] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/27/2023] [Accepted: 02/08/2023] [Indexed: 04/12/2024] Open
Abstract
The amyloid concept of Alzheimer's disease (AD) assumes the β-amyloid peptide (Aβ) as the main pathogenic factor, which injures neural and other brain cells, causing their malfunction and death. Although Aβ has been documented to exert its cytotoxic effect in a solitary manner, there is much evidence to claim that its toxicity can be modulated by other proteins. The list of such Aβ co-factors or interactors includes tau, APOE, transthyretin, and others. These molecules interact with the peptide and affect the ability of Aβ to form oligomers or aggregates, modulating its toxicity. Thus, the list of potential substances able to reduce the harmful effects of the peptide should include ones that can prevent the pathogenic interactions by specifically binding Aβ and/or its partners. In the present review, we discuss the data on Aβ-based complexes in AD pathogenesis and on the compounds directly targeting Aβ or the destructors of its complexes with other polypeptides.
Collapse
Affiliation(s)
- Vladimir F. Lazarev
- Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
| | - Elizaveta A. Dutysheva
- Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
| | - Igor E. Kanunikov
- Biological Faculty, St. Petersburg State University, 199034 Saint Petersburg, Russia
| | - Irina V. Guzhova
- Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
| | - Boris A. Margulis
- Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
| |
Collapse
|
10
|
O’Day DH. Calmodulin and Amyloid Beta as Coregulators of Critical Events during the Onset and Progression of Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24021393. [PMID: 36674908 PMCID: PMC9863087 DOI: 10.3390/ijms24021393] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 01/12/2023] Open
Abstract
Calmodulin (CaM) and a diversity of CaM-binding proteins (CaMBPs) are involved in the onset and progression of Alzheimer's disease (AD). In the amyloidogenic pathway, AβPP1, BACE1 and PSEN-1 are all calcium-dependent CaMBPs as are the risk factor proteins BIN1 and TREM2. Ca2+/CaM-dependent protein kinase II (CaMKII) and calcineurin (CaN) are classic CaMBPs involved in memory and plasticity, two events impacted by AD. Coupled with these events is the production of amyloid beta monomers (Aβ) and oligomers (Aβo). The recent revelations that Aβ and Aβo each bind to both CaM and to a host of Aβ receptors that are also CaMBPs adds a new level of complexity to our understanding of the onset and progression of AD. Multiple Aβ receptors that are proven CaMBPs (e.g., NMDAR, PMCA) are involved in calcium homeostasis an early event in AD and other neurodegenerative diseases. Other CaMBPs that are Aβ receptors are AD risk factors while still others are involved in the amyloidogenic pathway. Aβ binding to receptors not only serves to control CaM's ability to regulate critical proteins, but it is also implicated in Aβ turnover. The complexity of the Aβ/CaM/CaMBP interactions is analyzed using two events: Aβ generation and NMDAR function. The interactions between Aβ, CaM and CaMBPs reveals a new level of complexity to critical events associated with the onset and progression of AD and may help to explain the failure to develop successful therapeutic treatments for the disease.
Collapse
Affiliation(s)
- Danton H. O’Day
- Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada;
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
| |
Collapse
|
11
|
Johnson NR, Wang ACJ, Coughlan C, Sillau S, Lucero E, Viltz L, Markham N, Allen C, Dhanasekaran AR, Chial HJ, Potter H. Imipramine and olanzapine block apoE4-catalyzed polymerization of Aβ and show evidence of improving Alzheimer’s disease cognition. Alzheimers Res Ther 2022; 14:88. [PMID: 35768831 PMCID: PMC9241285 DOI: 10.1186/s13195-022-01020-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/11/2022] [Indexed: 01/18/2023]
Abstract
Background The apolipoprotein E (APOE) ε4 allele confers the strongest risk for late-onset Alzheimer’s disease (AD) besides age itself, but the mechanisms underlying this risk are debated. One hypothesis supported by evidence from multiple labs is that apoE4 binds to the amyloid-β (Aβ) peptide and catalyzes its polymerization into neurotoxic oligomers and fibrils. Inhibiting this early step in the amyloid cascade may thereby reduce or prevent neurodegeneration and AD. Methods Using a design of experiments (DOE) approach, we developed a high-throughput assay to identify inhibitors of apoE4-catalyzed polymerization of Aβ into oligomers and fibrils. We used it to screen the NIH Clinical Collection of small molecule drugs tested previously in human clinical trials. We then evaluated the efficacy and cytotoxicity of the hit compounds in primary neuron models of apoE4-induced Aβ and phosphorylated tau aggregation. Finally, we performed retrospective analyses of the National Alzheimer’s Coordinating Center (NACC) clinical dataset, using Cox regression and Cox proportional hazards models to determine if the use of two FDA-approved hit compounds was associated with better cognitive scores (Mini-Mental State Exam), or improved AD clinical diagnosis, when compared with other medications of the same clinical indication. Results Our high-throughput screen identified eight blood-brain barrier (BBB)-permeable hit compounds that reduced apoE4-catalyzed Aβ oligomer and fibril formation in a dose-dependent manner. Five hit compounds were non-toxic toward cultured neurons and also reduced apoE4-promoted Aβ and tau neuropathology in a dose-dependent manner. Three of the five compounds were determined to be specific inhibitors of apoE4, whereas the other two compounds were Aβ or tau aggregation inhibitors. When prescribed to AD patients for their normal clinical indications, two of the apoE4 inhibitors, imipramine and olanzapine, but not other (non-hit) antipsychotic or antidepressant medications, were associated with improvements in cognition and clinical diagnosis, especially among APOE4 carriers. Conclusions The critical test of any proposed AD mechanism is whether it leads to effective treatments. Our high-throughput screen identified two promising FDA-approved drugs, imipramine and olanzapine, which have no structural, functional, or clinical similarities other than their shared ability to inhibit apoE4-catalyzed Aβ polymerization, thus identifying this mechanism as an essential contribution of apoE4 to AD. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-022-01020-9.
Collapse
|
12
|
Wang HM, Seo CD, Lee KJ, Park JH, Lim HS. Evaluation of the cell permeability of bicyclic peptoids and bicyclic peptide-peptoid hybrids. Bioorg Chem 2022; 127:105976. [DOI: 10.1016/j.bioorg.2022.105976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 11/02/2022]
|
13
|
Moon HJ, Haroutunian V, Zhao L. Human apolipoprotein E isoforms are differentially sialylated and the sialic acid moiety in ApoE2 attenuates ApoE2-Aβ interaction and Aβ fibrillation. Neurobiol Dis 2022; 164:105631. [PMID: 35041991 PMCID: PMC9809161 DOI: 10.1016/j.nbd.2022.105631] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 01/05/2023] Open
Abstract
The APOE genotype is the most prominent genetic risk factor for the development of late-onset Alzheimer''s disease (LOAD); however, the underlying mechanisms remain unclear. In the present study, we found that the sialylation profiles of ApoE protein in the human brain are significantly different among the three isoforms, with ApoE2 exhibiting the most abundant sialic acid modification whereas ApoE4 had the least. We further observed that the sialic acid moiety in ApoE2 significantly affected the interaction between ApoE2 and Aβ peptides. The removal of sialic acid in ApoE2 increased the ApoE2 binding affinity for the Aβ17-24 region of Aβ and promoted Aβ fibrillation. These findings provide a plausible explanation for the well-documented differential roles of ApoE isoforms in Aβ pathogenesis. Specifically, compared to the other two isotypes, the higher expression of sialic acid in ApoE2 may contribute to the less potent interaction between ApoE2 and Aβ and ultimately the slower rate of brain Aβ deposition, a mechanism thought to underlie ApoE2-mediated decreased risk for AD. Future studies are warranted to determine whether the differential sialylation in ApoE isoforms may also contribute to some of their other distinct properties, such as their divergent preferences in associations with lipids and lipoproteins, as well as their potential impact on neuroinflammation through modulation of microglial Siglec activity. Overall, our findings lead to the insight that the sialic acid structure is an important posttranslational modification (PTM) that alters ApoE protein functions with relevance for AD.
Collapse
Affiliation(s)
- Hee-Jung Moon
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Vahram Haroutunian
- The Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 100029, USA
| | - Liqin Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA; Neuroscience Graduate Program, University of Kansas, Lawrence, KS 66045, USA.
| |
Collapse
|
14
|
Chai AB, Lam HHJ, Kockx M, Gelissen IC. Apolipoprotein E isoform-dependent effects on the processing of Alzheimer's amyloid-β. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158980. [PMID: 34044125 DOI: 10.1016/j.bbalip.2021.158980] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/16/2021] [Accepted: 05/20/2021] [Indexed: 12/28/2022]
Abstract
Since the identification of the apolipoprotein E (apoE) *ε4 allele as a major genetic risk factor for late-onset Alzheimer's disease, significant efforts have been aimed at elucidating how apoE4 expression confers greater brain amyloid-β (Aβ) burden, earlier disease onset and worse clinical outcomes compared to apoE2 and apoE3. ApoE primarily functions as a lipid carrier to regulate cholesterol metabolism in circulation as well as in the brain. However, it has also been suggested to interact with hydrophobic Aβ peptides to influence their processing in an isoform-dependent manner. Here, we review evidence from in vitro and in vivo studies extricating the effects of the three apoE isoforms, on different stages of the Aβ processing pathway including synthesis, aggregation, deposition, clearance and degradation. ApoE4 consistently correlates with impaired Aβ clearance, however data regarding Aβ synthesis and aggregation are conflicting and likely reflect inconsistencies in experimental approaches across studies. We further discuss the physical and chemical properties of apoE that may explain the inherent differences in activity between the isoforms. The lipidation status and lipid transport function of apoE are intrinsically linked with its ability to interact with Aβ. Traditionally, apoE-oriented therapeutic strategies for Alzheimer's disease have been proposed to non-specifically enhance or inhibit apoE activity. However, given the wide-ranging physiological functions of apoE in the brain and periphery, a more viable approach may be to specifically target and neutralise the pathological apoE4 isoform.
Collapse
Affiliation(s)
- Amanda B Chai
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Hin Hei Julian Lam
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Maaike Kockx
- ANZAC Research Institute, Concord Repatriation General Hospital, University of Sydney, Concord, NSW 2139, Australia
| | - Ingrid C Gelissen
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
15
|
Roda AR, Montoliu-Gaya L, Villegas S. The Role of Apolipoprotein E Isoforms in Alzheimer's Disease. J Alzheimers Dis 2020; 68:459-471. [PMID: 30775980 DOI: 10.3233/jad-180740] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD), the most common type of dementia worldwide, is characterized by high levels of amyloid-β (Aβ) peptide and hyperphosphorylated tau protein. Genetically, the ɛ4 allele of apolipoprotein E (ApoE) has been established as the major risk factor for developing late-onset AD (LOAD), the most common form of the disease. Although the role ApoE plays in AD is still not completely understood, a differential role of its isoforms has long been known. The current review compiles the involvement of ApoE isoforms in amyloid-β protein precursor transcription, Aβ aggregation and clearance, synaptic plasticity, neuroinflammation, lipid metabolism, mitochondrial function, and tau hyperphosphorylation. Due to the complexity of LOAD, an accurate description of the interdependence among all the related molecular mechanisms involved in the disease is needed for developing successful therapies.
Collapse
Affiliation(s)
- Alejandro R Roda
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Laia Montoliu-Gaya
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.,Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Sandra Villegas
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| |
Collapse
|
16
|
Gatti L, Tinelli F, Scelzo E, Arioli F, Di Fede G, Obici L, Pantoni L, Giaccone G, Caroppo P, Parati EA, Bersano A. Understanding the Pathophysiology of Cerebral Amyloid Angiopathy. Int J Mol Sci 2020; 21:ijms21103435. [PMID: 32414028 PMCID: PMC7279405 DOI: 10.3390/ijms21103435] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/04/2020] [Accepted: 05/08/2020] [Indexed: 12/12/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA), one of the main types of cerebral small vessel disease, is a major cause of spontaneous intracerebral haemorrhage and an important contributor to cognitive decline in elderly patients. Despite the number of experimental in vitro studies and animal models, the pathophysiology of CAA is still largely unknown. Although several pathogenic mechanisms including an unbalance between production and clearance of amyloid beta (Aβ) protein as well as ‘the prion hypothesis’ have been invoked as possible disease triggers, they do not explain completely the disease pathogenesis. This incomplete disease knowledge limits the implementation of treatments able to prevent or halt the clinical progression. The continuous increase of CAA patients makes imperative the development of suitable experimental in vitro or animal models to identify disease biomarkers and new pharmacological treatments that could be administered in the early disease stages to prevent irreversible changes and disease progression.
Collapse
Affiliation(s)
- Laura Gatti
- Neurobiology Laboratory, Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (L.G.); (F.T.); (F.A.)
| | - Francesca Tinelli
- Neurobiology Laboratory, Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (L.G.); (F.T.); (F.A.)
| | - Emma Scelzo
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (E.S.); (E.A.P.)
| | - Francesco Arioli
- Neurobiology Laboratory, Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (L.G.); (F.T.); (F.A.)
| | - Giuseppe Di Fede
- Unit of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (G.D.F.); (G.G.); (P.C.)
| | - Laura Obici
- Amyloidosis Research and Treatment Centre, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Leonardo Pantoni
- “Luigi Sacco” Department of Biomedical and Clinical Sciences, University of Milan, 20157 Milan, Italy;
| | - Giorgio Giaccone
- Unit of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (G.D.F.); (G.G.); (P.C.)
| | - Paola Caroppo
- Unit of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (G.D.F.); (G.G.); (P.C.)
| | - Eugenio Agostino Parati
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (E.S.); (E.A.P.)
| | - Anna Bersano
- Cerebrovascular Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy; (E.S.); (E.A.P.)
- Correspondence: ; Tel.: +39-0223943310
| |
Collapse
|
17
|
Wisniewski T, Drummond E. APOE-amyloid interaction: Therapeutic targets. Neurobiol Dis 2020; 138:104784. [PMID: 32027932 PMCID: PMC7118587 DOI: 10.1016/j.nbd.2020.104784] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/22/2020] [Accepted: 01/31/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder that is growing in prevalence globally. It is the only major cause of death without any effective pharmacological means to treat or slow progression. Inheritance of the ε4 allele of the Apolipoprotein (APO) E gene is the strongest genetic risk factor for late-onset AD. The interaction between APOE and amyloid β (Aβ) plays a key role in AD pathogenesis. The APOE-Aβ interaction regulates Aβ aggregation and clearance and therefore directly influences the development of amyloid plaques, congophilic amyloid angiopathy and subsequent tau related pathology. Relatively few AD therapeutic approaches have directly targeted the APOE-Aβ interaction thus far. Here we review the critical role of APOE in the pathogenesis of AD and some of the most promising therapeutic approaches that focus on the APOE-Aβ interaction.
Collapse
Affiliation(s)
- Thomas Wisniewski
- Departments of Neurology, Pathology and Psychiatry, Center for Cognitive Neurology, NYU School of Medicine, Science Building, Rm 1017, 435 East 30(th) Street, New York, NY 10016, USA.
| | - Eleanor Drummond
- Brain & Mind Centre and Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
18
|
Iacono D, Feltis GC. Impact of Apolipoprotein E gene polymorphism during normal and pathological conditions of the brain across the lifespan. Aging (Albany NY) 2020; 11:787-816. [PMID: 30677746 PMCID: PMC6366964 DOI: 10.18632/aging.101757] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 01/05/2019] [Indexed: 12/12/2022]
Abstract
The central nervous system (CNS) is the cellular substrate for the integration of complex, dynamic, constant, and simultaneous interactions among endogenous and exogenous stimuli across the entire human lifespan. Numerous studies on aging-related brain diseases show that some genes identified as risk factors for some of the most common neurodegenerative diseases - such as the allele 4 of APOE gene (APOE4) for Alzheimer's disease (AD) - have a much earlier neuro-anatomical and neuro-physiological impact. The impact of APOE polymorphism appears in fact to start as early as youth and early-adult life. Intriguingly, though, those same genes associated with aging-related brain diseases seem to influence different aspects of the brain functioning much earlier actually, that is, even from the neonatal periods and earlier. The APOE4, an allele classically associated with later-life neurodegenerative disorders as AD, seems in fact to exert a series of very early effects on phenomena of neuroplasticity and synaptogenesis that begin from the earliest periods of life such as the fetal ones.We reviewed some of the findings supporting the hypothesis that APOE polymorphism is an early modifier of various neurobiological aspects across the entire human lifespan - from the in-utero to the centenarian life - during both normal and pathological conditions of the brain.
Collapse
Affiliation(s)
- Diego Iacono
- Neuropathology Research, Biomedical Research Institute of New Jersey (BRInj), Cedar Knolls, NJ 07927, USA.,MidAtlantic Neonatology Associates (MANA), Morristown, NJ 07960, USA.,Atlantic Neuroscience Institute, Atlantic Health System (AHS), Overlook Medical Center, Summit, NJ 07901, USA
| | - Gloria C Feltis
- Neuropathology Research, Biomedical Research Institute of New Jersey (BRInj), Cedar Knolls, NJ 07927, USA
| |
Collapse
|
19
|
Bhute S, Sarmah D, Datta A, Rane P, Shard A, Goswami A, Borah A, Kalia K, Dave KR, Bhattacharya P. Molecular Pathogenesis and Interventional Strategies for Alzheimer's Disease: Promises and Pitfalls. ACS Pharmacol Transl Sci 2020; 3:472-488. [PMID: 32566913 DOI: 10.1021/acsptsci.9b00104] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is a debilitating disorder characterized by age-related dementia, which has no effective treatment to date. β-Amyloid depositions and hyperphosphorylated tau proteins are the main pathological hallmarks, along with oxidative stress, N-methyl-d-aspartate (NMDA) receptor-mediated excitotoxicity, and low levels of acetylcholine. Current pharmacotherapy for AD only provides symptomatic relief and limited improvement in cognitive functions. Many molecules have been explored that show promising outcomes in AD therapy and can regulate cellular survival through different pathways. To have a vivid approach to strategize the treatment regimen, AD physiopathology should be better explained considering diverse etiological factors in conjunction with biochemical disturbances. This Review attempts to discuss different disease modification approaches and address the novel therapeutic targets of AD that might pave the way for new drug discovery using the well-defined targets for therapy of the disease.
Collapse
Affiliation(s)
- Shashikala Bhute
- Department of Pharmacology and Toxicology,National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382355, Gujarat, India
| | - Deepaneeta Sarmah
- Department of Pharmacology and Toxicology,National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382355, Gujarat, India
| | - Aishika Datta
- Department of Pharmacology and Toxicology,National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382355, Gujarat, India
| | - Pallavi Rane
- Department of Pharmacology and Toxicology,National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382355, Gujarat, India
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382355, Gujarat, India
| | - Avirag Goswami
- Department of Neurology, Albert Einstein Medical Center, Philadelphia, Pennsylvania 19141, United States
| | - Anupom Borah
- Department of Life Science and Bioinformatics, Assam University, Silchar, Assam-788011, India
| | - Kiran Kalia
- Department of Pharmacology and Toxicology,National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382355, Gujarat, India
| | - Kunjan R Dave
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology,National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar-382355, Gujarat, India
| |
Collapse
|
20
|
Cotrina EY, Gimeno A, Llop J, Jiménez-Barbero J, Quintana J, Valencia G, Cardoso I, Prohens R, Arsequell G. Calorimetric Studies of Binary and Ternary Molecular Interactions between Transthyretin, Aβ Peptides, and Small-Molecule Chaperones toward an Alternative Strategy for Alzheimer's Disease Drug Discovery. J Med Chem 2020; 63:3205-3214. [PMID: 32124607 PMCID: PMC7115756 DOI: 10.1021/acs.jmedchem.9b01970] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
![]()
Transthyretin
(TTR) modulates the deposition, processing, and toxicity
of Abeta (Aβ) peptides. We have shown that this effect is enhanced
in mice by treatment with small molecules such as iododiflunisal (IDIF, 4), a good TTR stabilizer. Here, we describe the thermodynamics
of the formation of binary and ternary complexes among TTR, Aβ(1–42)
peptide, and TTR stabilizers using isothermal titration calorimetry
(ITC). A TTR/Aβ(1–42) (1:1)
complex with a dissociation constant of Kd = 0.94 μM is formed; with IDIF
(4), this constant improves up to Kd = 0.32 μM, indicating
the presence of a ternary complex TTR/IDIF/Aβ(1–42).
However, with the drugs diflunisal (1) or Tafamidis (2), an analogous chaperoning effect could not be observed.
Similar phenomena could be recorded with the shorter peptide Aβ(12–28)
(7). We propose the design of a simple assay system for
the search of other chaperones that behave like IDIF and may become
potential candidate drugs for Alzheimer’s disease (AD).
Collapse
Affiliation(s)
- Ellen Y Cotrina
- Institut de Quı́mica Avançada de Catalunya (I.Q.A.C.-C.S.I.C.), 08034 Barcelona, Spain
| | - Ana Gimeno
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 800, 48160 Derio, Spain
| | - Jordi Llop
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 San Sebastian, Spain
| | - Jesús Jiménez-Barbero
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 800, 48160 Derio, Spain.,Ikerbasque, Basque Foundation for Science, Maria Diaz de Haro 13, 48009 Bilbao, Spain.,Department of Organic Chemistry II, Faculty of Science & Technology, University of the Basque Country, 48940 Leioa, Bizkaia, Spain
| | - Jordi Quintana
- Research Programme on Biomedical Informatics, Universitat Pompeu Fabra (UPF-IMIM), 08003 Barcelona, Spain
| | - Gregorio Valencia
- Institut de Quı́mica Avançada de Catalunya (I.Q.A.C.-C.S.I.C.), 08034 Barcelona, Spain
| | - Isabel Cardoso
- IBMC-Instituto de Biologia Molecular e Celular, 4200-135 Porto, Portugal.,i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Rafel Prohens
- Unitat de Polimorfisme i Calorimetria, Centres Cientı́fics i Tecnològics, Universitat de Barcelona, Baldiri Reixac 10, 08028 Barcelona, Spain
| | - Gemma Arsequell
- Institut de Quı́mica Avançada de Catalunya (I.Q.A.C.-C.S.I.C.), 08034 Barcelona, Spain
| |
Collapse
|
21
|
Armiento V, Spanopoulou A, Kapurniotu A. Peptide-Based Molecular Strategies To Interfere with Protein Misfolding, Aggregation, and Cell Degeneration. Angew Chem Int Ed Engl 2020; 59:3372-3384. [PMID: 31529602 PMCID: PMC7064928 DOI: 10.1002/anie.201906908] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Indexed: 12/31/2022]
Abstract
Protein misfolding into amyloid fibrils is linked to more than 40 as yet incurable cell- and neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and type 2 diabetes. So far, however, only one of the numerous anti-amyloid molecules has reached patients. This Minireview gives an overview of molecular strategies and peptide chemistry "tools" to design, develop, and discover peptide-based molecules as anti-amyloid drug candidates. We focus on two major inhibitor rational design strategies: 1) the oldest and most common strategy, based on molecular recognition elements of amyloid self-assembly, and 2) a more recent approach, based on cross-amyloid interactions. We discuss why peptide-based amyloid inhibitors, in particular their advanced generations, can be promising leads or candidates for anti-amyloid drugs as well as valuable tools for deciphering amyloid-mediated cell damage and its link to disease pathogenesis.
Collapse
Affiliation(s)
- Valentina Armiento
- Division of Peptide BiochemistryTUM School of Life SciencesTechnische Universität MünchenEmil-Erlenmeyer-Forum 585354FreisingGermany
| | - Anna Spanopoulou
- Division of Peptide BiochemistryTUM School of Life SciencesTechnische Universität MünchenEmil-Erlenmeyer-Forum 585354FreisingGermany
- Current address: Coriolis Pharma Research GmbHFraunhoferstrasse 18B82152PlaneggGermany
| | - Aphrodite Kapurniotu
- Division of Peptide BiochemistryTUM School of Life SciencesTechnische Universität MünchenEmil-Erlenmeyer-Forum 585354FreisingGermany
| |
Collapse
|
22
|
Lewandowski CT, Maldonado Weng J, LaDu MJ. Alzheimer's disease pathology in APOE transgenic mouse models: The Who, What, When, Where, Why, and How. Neurobiol Dis 2020; 139:104811. [PMID: 32087290 DOI: 10.1016/j.nbd.2020.104811] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/01/2020] [Accepted: 02/18/2020] [Indexed: 02/07/2023] Open
Abstract
The focus on amyloid plaques and neurofibrillary tangles has yielded no Alzheimer's disease (AD) modifying treatments in the past several decades, despite successful studies in preclinical mouse models. This inconsistency has caused a renewed focus on improving the fidelity and reliability of AD mouse models, with disparate views on how this improvement can be accomplished. However, the interactive effects of the universal biological variables of AD, which include age, APOE genotype, and sex, are often overlooked. Age is the greatest risk factor for AD, while the ε4 allele of the human APOE gene, encoding apolipoprotein E, is the greatest genetic risk factor. Sex is the final universal biological variable of AD, as females develop AD at almost twice the rate of males and, importantly, female sex exacerbates the effects of APOE4 on AD risk and rate of cognitive decline. Therefore, this review evaluates the importance of context for understanding the role of APOE in preclinical mouse models. Specifically, we detail how human AD pathology is mirrored in current transgenic mouse models ("What") and describe the critical need for introducing human APOE into these mouse models ("Who"). We next outline different methods for introducing human APOE into mice ("How") and highlight efforts to develop temporally defined and location-specific human apoE expression models ("When" and "Where"). We conclude with the importance of choosing the human APOE mouse model relevant to the question being addressed, using the selection of transgenic models for testing apoE-targeted therapeutics as an example ("Why").
Collapse
Affiliation(s)
- Cutler T Lewandowski
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, 833 S. Wood St., Chicago, IL 60612, USA.
| | - Juan Maldonado Weng
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 S. Wood St., Chicago, IL 60612, USA.
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 S. Wood St., Chicago, IL 60612, USA.
| |
Collapse
|
23
|
Butterfield DA, Mattson MP. Apolipoprotein E and oxidative stress in brain with relevance to Alzheimer's disease. Neurobiol Dis 2020; 138:104795. [PMID: 32036033 DOI: 10.1016/j.nbd.2020.104795] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/26/2020] [Accepted: 02/06/2020] [Indexed: 02/08/2023] Open
Abstract
Inheritance of apolipoprotein E4 (APOE4) is a major risk factor for development of Alzheimer's disease (AD). This lipoprotein, in contrast to apoE2, has arginine residues at positions 112 and 158 in place of cysteines in the latter isoform. In apoE3, the Cys at residue 158 is replaced by an arginine residue. This differential amino acid composition of the three genotypes of APOE have profound influence on the structure, binding properties, and multiple functions of this lipoprotein. Moreover, AD brain is under a high degree of oxidative stress, including that associated with amyloid β-peptide (Aβ) oligomers. Lipid peroxidation produces the highly reactive and neurotoxic molecule, 4-hydroxynonenal (HNE) that forms covalent bonds with cysteine residues (Cys) [as well as with Lys and His residues]. Covalently modified Cys significantly alter structure and function of modified proteins. HNE bound to Cys residue(s) on apoE2 and apoE3 lessens the chance of HNE damage other proteins. apoE4, lacking Cys residues, is unable to scavenge HNE, permitting this latter neurotoxic molecule to lead to oxidative modification of neuronal proteins and eventual cell death. We posit that this lack of HNE scavenging activity in apoE4 significantly contributes to the association of APOE4 inheritance and increased risk of developing AD. Apoe knock-out mice provide insights into the role of this lipoprotein in oxidative stress. Targeted replacement mice in which the mouse gene of Apoe is separately replaced by the human APOE2, APOE3, or APOE4 genes, while keeping the mouse promoter assures the correct location and amount of the human protein isoform. Human APOE targeted replacement mice have been used to investigate the notion that oxidative damage to and death of neurons in AD and its earlier stages is related to APOE genotype. This current paper reviews the intersection of human APOE genotype, oxidative stress, and diminished function of this lipoprotein as a major contributing risk factor for development of AD. Discussion of potential therapeutic strategies to mitigate against the elevated risk of developing AD with inheritance of the APOE4 allele also is presented.
Collapse
Affiliation(s)
- D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA.
| | - Mark P Mattson
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
24
|
Gharibyan AL, Islam T, Pettersson N, Golchin SA, Lundgren J, Johansson G, Genot M, Schultz N, Wennström M, Olofsson A. Apolipoprotein E Interferes with IAPP Aggregation and Protects Pericytes from IAPP-Induced Toxicity. Biomolecules 2020; 10:biom10010134. [PMID: 31947546 PMCID: PMC7022431 DOI: 10.3390/biom10010134] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/10/2020] [Accepted: 01/12/2020] [Indexed: 02/06/2023] Open
Abstract
Apolipoprotein E (ApoE) has become a primary focus of research after the discovery of its strong linkage to Alzheimer’s disease (AD), where the ApoE4 variant is the highest genetic risk factor for this disease. ApoE is commonly found in amyloid deposits of different origins, and its interaction with amyloid-β peptide (Aβ), the hallmark of AD, is well known. However, studies on the interaction of ApoEs with other amyloid-forming proteins are limited. Islet amyloid polypeptide (IAPP) is an amyloid-forming peptide linked to the development of type-2 diabetes and has also been shown to be involved in AD pathology and vascular dementia. Here we studied the impact of ApoE on IAPP aggregation and IAPP-induced toxicity on blood vessel pericytes. Using both in vitro and cell-based assays, we show that ApoE efficiently inhibits the amyloid formation of IAPP at highly substoichiometric ratios and that it interferes with both nucleation and elongation. We also show that ApoE protects the pericytes against IAPP-induced toxicity, however, the ApoE4 variant displays the weakest protective potential. Taken together, our results suggest that ApoE has a generic amyloid-interfering property and can be protective against amyloid-induced cytotoxicity, but there is a loss of function for the ApoE4 variant.
Collapse
Affiliation(s)
- Anna L. Gharibyan
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-901 87 Umeå, Sweden; (T.I.); (N.P.); (S.A.G.); (J.L.); (G.J.); (M.G.)
- Correspondence: (A.L.G.); (A.O.); Tel.: +46-73-912-54-94 (A.L.G.); +46-70-354-33-01 (A.O.)
| | - Tohidul Islam
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-901 87 Umeå, Sweden; (T.I.); (N.P.); (S.A.G.); (J.L.); (G.J.); (M.G.)
| | - Nina Pettersson
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-901 87 Umeå, Sweden; (T.I.); (N.P.); (S.A.G.); (J.L.); (G.J.); (M.G.)
| | - Solmaz A. Golchin
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-901 87 Umeå, Sweden; (T.I.); (N.P.); (S.A.G.); (J.L.); (G.J.); (M.G.)
| | - Johanna Lundgren
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-901 87 Umeå, Sweden; (T.I.); (N.P.); (S.A.G.); (J.L.); (G.J.); (M.G.)
| | - Gabriella Johansson
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-901 87 Umeå, Sweden; (T.I.); (N.P.); (S.A.G.); (J.L.); (G.J.); (M.G.)
| | - Mélany Genot
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-901 87 Umeå, Sweden; (T.I.); (N.P.); (S.A.G.); (J.L.); (G.J.); (M.G.)
| | - Nina Schultz
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 21428 Malmö, Sweden; (N.S.); (M.W.)
| | - Malin Wennström
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, 21428 Malmö, Sweden; (N.S.); (M.W.)
| | - Anders Olofsson
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-901 87 Umeå, Sweden; (T.I.); (N.P.); (S.A.G.); (J.L.); (G.J.); (M.G.)
- Correspondence: (A.L.G.); (A.O.); Tel.: +46-73-912-54-94 (A.L.G.); +46-70-354-33-01 (A.O.)
| |
Collapse
|
25
|
Armiento V, Spanopoulou A, Kapurniotu A. Peptid‐basierte molekulare Strategien zum Einsatz bei Proteinfehlfaltung, Proteinaggregation und Zelldegeneration. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201906908] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Valentina Armiento
- Fachgebiet PeptidbiochemieTUM School of Life SciencesTechnische Universität München Emil-Erlenmeyer-Forum 5 85354 Freising Deutschland
| | - Anna Spanopoulou
- Fachgebiet PeptidbiochemieTUM School of Life SciencesTechnische Universität München Emil-Erlenmeyer-Forum 5 85354 Freising Deutschland
- Aktuelle Adresse: Coriolis Pharma Research GmbH Fraunhoferstraße 18B 82152 Planegg Deutschland
| | - Aphrodite Kapurniotu
- Fachgebiet PeptidbiochemieTUM School of Life SciencesTechnische Universität München Emil-Erlenmeyer-Forum 5 85354 Freising Deutschland
| |
Collapse
|
26
|
Islam T, Gharibyan AL, Golchin SA, Pettersson N, Brännström K, Hedberg I, Virta MM, Olofsson L, Olofsson A. Apolipoprotein E impairs amyloid-β fibril elongation and maturation. FEBS J 2019; 287:1208-1219. [PMID: 31571352 DOI: 10.1111/febs.15075] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/18/2019] [Accepted: 09/27/2019] [Indexed: 01/01/2023]
Abstract
Alzheimer's disease (AD) is strongly linked to amyloid depositions of the Aβ peptide (Aβ). The lipid-binding protein apolipoprotein E (ApoE) has been found to interfere with Aβ amyloid formation and to exert a strong clinical impact to the pathology of AD. The APOE gene exists in three allelic isoforms represented by APOE ε2, APOE ε3, and APOE ε4. Carriers of the APOE ε4 variant display a gene dose-dependent increased risk of developing the disease. Aβ amyloids are formed via a nucleation-dependent mechanism where free monomers are added onto a nucleus in a template-dependent manner. Using a combination of surface plasmon resonance and thioflavin-T assays, we here show that ApoE can target the process of fibril elongation and that its interference effectively prevents amyloid maturation. We expose a complex equilibrium where the concentration of ApoE, Aβ monomers, and the amount of already formed Aβ fibrils will affect the relative proportion and formation rate of mature amyloids versus alternative assemblies. The result illustrates a mechanism which may affect both the clearance rate of Aβ assemblies in vivo and the population of cytotoxic Aβ assemblies.
Collapse
Affiliation(s)
- Tohidul Islam
- Department of Medical Biochemistry and Biophysics, Umeå University, Sweden
| | - Anna L Gharibyan
- Department of Medical Biochemistry and Biophysics, Umeå University, Sweden
| | - Solmaz A Golchin
- Department of Medical Biochemistry and Biophysics, Umeå University, Sweden
| | - Nina Pettersson
- Department of Medical Biochemistry and Biophysics, Umeå University, Sweden
| | | | - Isabell Hedberg
- Department of Medical Biochemistry and Biophysics, Umeå University, Sweden
| | - Merit-Miriam Virta
- Department of Medical Biochemistry and Biophysics, Umeå University, Sweden
| | - Linnea Olofsson
- Department of Medical Biochemistry and Biophysics, Umeå University, Sweden
| | - Anders Olofsson
- Department of Medical Biochemistry and Biophysics, Umeå University, Sweden
| |
Collapse
|
27
|
Rios X, Gómez-Vallejo V, Martín A, Cossío U, Morcillo MÁ, Alemi M, Cardoso I, Quintana J, Jiménez-Barbero J, Cotrina EY, Valencia G, Arsequell G, Llop J. Radiochemical examination of transthyretin (TTR) brain penetration assisted by iododiflunisal, a TTR tetramer stabilizer and a new candidate drug for AD. Sci Rep 2019; 9:13672. [PMID: 31541162 PMCID: PMC6754432 DOI: 10.1038/s41598-019-50071-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 08/31/2019] [Indexed: 12/21/2022] Open
Abstract
It is well settled that the amyloidogenic properties of the plasma protein transporter transthyretin (TTR) can be modulated by compounds that stabilize its native tetrameric conformation. TTR is also present in cerebrospinal fluid where it can bind to Aβ-peptides and prevent Aβ aggregation. We have previously shown that treatment of Alzheimer’s Disease (AD) model mice with iododiflunisal (IDIF), a TTR tetramer stabilizing compound, prevents AD pathologies. This evidence positioned IDIF as a new lead drug for AD. In dissecting the mechanism of action of IDIF, we disclose here different labeling strategies for the preparation of 131I-labeled IDIF and 131I- and 124I-labeled TTR, which have been further used for the preparation of IDIF-TTR complexes labeled either on the compound or the protein. The biodistribution of all labeled species after intravenous administration has been investigated in mice using ex vivo and in vivo techniques. Our results confirm the capacity of TTR to cross the blood brain barrier (BBB) and suggest that the formation of TTR-IDIF complexes enhances BBB permeability of both IDIF and TTR. The increased TTR and IDIF brain concentrations may result in higher Aβ-peptide sequestration capacity with the subsequent inhibition of AD symptoms as we have previously observed in mice.
Collapse
Affiliation(s)
- Xabier Rios
- Radiochemistry and Nuclear Imaging Group, CIC biomaGUNE, 20014, San Sebastián, Guipúzcoa, Spain
| | - Vanessa Gómez-Vallejo
- Radiochemistry and Nuclear Imaging Group, CIC biomaGUNE, 20014, San Sebastián, Guipúzcoa, Spain
| | - Abraham Martín
- Achucarro Basque Center for Neuroscience, 48940, Leioa, Spain.,Ikerbasque Basque Foundation for Science, Maria Díaz de Haro 3, 48013, Bilbao, Spain
| | - Unai Cossío
- Radiochemistry and Nuclear Imaging Group, CIC biomaGUNE, 20014, San Sebastián, Guipúzcoa, Spain
| | - Miguel Ángel Morcillo
- Biomedical Applications of Radioisotopes and Pharmacokinetics Unit, CIEMAT, 28040, Madrid, Spain
| | - Mobina Alemi
- IBMC - Instituto de Biologia Celular e Molecular, i3S-Instituto de Investigação e Inovação em Saúde, 4200-135, Porto, Portugal
| | - Isabel Cardoso
- IBMC - Instituto de Biologia Celular e Molecular, i3S-Instituto de Investigação e Inovação em Saúde, 4200-135, Porto, Portugal
| | - Jordi Quintana
- Plataforma Drug Discovery, Parc Científic de Barcelona (PCB), 08028, Barcelona, Spain.,Research Programme on Biomedical Informatics, Universitat Pompeu Fabra, 08003, Barcelona, Spain
| | - Jesús Jiménez-Barbero
- Ikerbasque Basque Foundation for Science, Maria Díaz de Haro 3, 48013, Bilbao, Spain.,CIC bioGUNE, Bizkaia Technology Park, Building 800, 48160, Derio, Spain
| | - Ellen Y Cotrina
- Institut de Química Avançada de Catalunya (I.Q.A.C.-C.S.I.C.), 08034, Barcelona, Spain
| | - Gregorio Valencia
- Institut de Química Avançada de Catalunya (I.Q.A.C.-C.S.I.C.), 08034, Barcelona, Spain
| | - Gemma Arsequell
- Institut de Química Avançada de Catalunya (I.Q.A.C.-C.S.I.C.), 08034, Barcelona, Spain.
| | - Jordi Llop
- Radiochemistry and Nuclear Imaging Group, CIC biomaGUNE, 20014, San Sebastián, Guipúzcoa, Spain. .,Centro de Investigación Biomédica en Red- Enfermedades Respiratorias (CIBERES), Madrid, Spain.
| |
Collapse
|
28
|
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: 3.3] [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
|
29
|
The role of APOE4 in Alzheimer's disease: strategies for future therapeutic interventions. Neuronal Signal 2019; 3:NS20180203. [PMID: 32269835 PMCID: PMC7104324 DOI: 10.1042/ns20180203] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 03/25/2019] [Accepted: 03/27/2019] [Indexed: 12/25/2022] Open
Abstract
Alzheimer’s disease (AD) is the leading cause of dementia affecting almost 50 million people worldwide. The ε4 allele of Apolipoprotein E (APOE) is the strongest known genetic risk factor for late-onset AD cases, with homozygous APOE4 carriers being approximately 15-times more likely to develop the disease. With 25% of the population being APOE4 carriers, understanding the role of this allele in AD pathogenesis and pathophysiology is crucial. Though the exact mechanism by which ε4 allele increases the risk for AD is unknown, the processes mediated by APOE, including cholesterol transport, synapse formation, modulation of neurite outgrowth, synaptic plasticity, destabilization of microtubules, and β-amyloid clearance, suggest potential therapeutic targets. This review will summarize the impact of APOE on neurons and neuronal signaling, the interactions between APOE and AD pathology, and the association with memory decline. We will then describe current treatments targeting APOE4, complications associated with the current therapies, and suggestions for future areas of research and treatment.
Collapse
|
30
|
Boutajangout A, Lindberg H, Awwad A, Paul A, Baitalmal R, Almokyad I, Höidén-Guthenberg I, Gunneriusson E, Frejd FY, Härd T, Löfblom J, Ståhl S, Wisniewski T. Affibody-Mediated Sequestration of Amyloid β Demonstrates Preventive Efficacy in a Transgenic Alzheimer's Disease Mouse Model. Front Aging Neurosci 2019; 11:64. [PMID: 30967771 PMCID: PMC6440316 DOI: 10.3389/fnagi.2019.00064] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/06/2019] [Indexed: 12/21/2022] Open
Abstract
Different strategies for treatment and prevention of Alzheimer’s disease (AD) are currently under investigation, including passive immunization with anti-amyloid β (anti-Aβ) monoclonal antibodies (mAbs). Here, we investigate the therapeutic potential of a novel type of Aβ-targeting agent based on an affibody molecule with fundamentally different properties to mAbs. We generated a therapeutic candidate, denoted ZSYM73-albumin-binding domain (ABD; 16.8 kDa), by genetic linkage of the dimeric ZSYM73 affibody for sequestering of monomeric Aβ-peptides and an ABD for extension of its in vivo half-life. Amyloid precursor protein (APP)/PS1 transgenic AD mice were administered with ZSYM73-ABD, followed by behavioral examination and immunohistochemistry. Results demonstrated rescued cognitive functions and significantly lower amyloid burden in the treated animals compared to controls. No toxicological symptoms or immunology-related side-effects were observed. To our knowledge, this is the first reported in vivo investigation of a systemically delivered scaffold protein against monomeric Aβ, demonstrating a therapeutic potential for prevention of AD.
Collapse
Affiliation(s)
- Allal Boutajangout
- Center for Cognitive Neurology, New York University Langone Health, New York, NY, United States.,Department of Neurology, New York University Langone Health, New York, NY, United States.,Department of Psychiatry, New York University Langone Health, New York, NY, United States.,Department of Physiology & Neuroscience, New York University Langone Medical Center, New York, NY, United States
| | - Hanna Lindberg
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Abdulaziz Awwad
- School of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Arun Paul
- Center for Cognitive Neurology, New York University Langone Health, New York, NY, United States.,Department of Neurology, New York University Langone Health, New York, NY, United States
| | - Rabaa Baitalmal
- Center for Cognitive Neurology, New York University Langone Health, New York, NY, United States.,Department of Neurology, New York University Langone Health, New York, NY, United States
| | - Ismail Almokyad
- Center for Cognitive Neurology, New York University Langone Health, New York, NY, United States.,Department of Neurology, New York University Langone Health, New York, NY, United States
| | | | | | | | - Torleif Härd
- Department of Chemistry and Biotechnology, Swedish University of Agricultural Sciences (SLU), Uppsala, Sweden
| | - John Löfblom
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Stefan Ståhl
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Thomas Wisniewski
- Center for Cognitive Neurology, New York University Langone Health, New York, NY, United States.,Department of Neurology, New York University Langone Health, New York, NY, United States.,Department of Psychiatry, New York University Langone Health, New York, NY, United States.,Department of Pathology, New York University School of Medicine, New York, NY, United States
| |
Collapse
|
31
|
Abstract
BACKGROUND The growing body of evidence indicating the heterogeneity of Alzheimer's disease (AD), coupled with disappointing clinical studies directed at a fit-for-all therapy, suggest that the development of a single magic cure suitable for all cases may not be possible. This calls for a shift in paradigm where targeted treatment is developed for specific AD subpopulations that share distinct genetic or pathological properties. Apolipoprotein E4 (apoE4), the most prevalent genetic risk factor of AD, is expressed in more than half of AD patients and is thus an important possible AD therapeutic target. REVIEW This review focuses initially on the pathological effects of apoE4 in AD, as well as on the corresponding cellular and animal models and the suggested cellular and molecular mechanisms which mediate them. The second part of the review focuses on recent apoE4-targeted (from the APOE gene to the apoE protein and its interactors) therapeutic approaches that have been developed in animal models and are ready to be translated to human. Further, the issue of whether the pathological effects of apoE4 are due to loss of protective function or due to gain of toxic function is discussed herein. It is possible that both mechanisms coexist, with certain constituents of the apoE4 molecule and/or its downstream signaling mediating a toxic effect, while others are associated with a loss of protective function. CONCLUSION ApoE4 is a promising AD therapeutic target that remains understudied. Recent studies are now paving the way for effective apoE4-directed AD treatment approaches.
Collapse
|
32
|
Pradhan K, Das G, Gupta V, Mondal P, Barman S, Khan J, Ghosh S. Discovery of Neuroregenerative Peptoid from Amphibian Neuropeptide That Inhibits Amyloid-β Toxicity and Crosses Blood-Brain Barrier. ACS Chem Neurosci 2019; 10:1355-1368. [PMID: 30408415 DOI: 10.1021/acschemneuro.8b00427] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Development of potential therapeutics for Alzheimer's disease (AD) requires a multifaceted strategy considering the high levels of complexity of the human brain and its mode of function. Here, we adopted an advanced strategy targeting two key pathological hallmarks of AD: senile plaques and neurofibrillary tangles. We derived a lead short tetrapeptide, Ser-Leu-Lys-Pro (SLKP), from a dodeca-neuropeptide of amphibian (frog) brain. Results suggested that the SLKP peptide had a superior effect compared to the dodecapeptide in neuroprotection. This result encouraged us to adopt peptidomimetic approach to synthesize an SLKP peptoid. Remarkably, we found that the SLKP peptoid is more potent than its peptide analogue, which significantly inhibits Aβ fibrillization, moderately binds with tubulin, and promotes tubulin polymerization as well as stabilization of microtubule networks. Further, we found that SLKP peptoid is stable in serum, shows significant neuroprotection against Aβ mediated toxicity, promotes significant neurite outgrowth, maintains healthy morphology of rat primary cortical neurons and crosses the blood-brain barrier (BBB). To the best of our knowledge, our SLKP peptoid is the first and shortest peptoid to show significant neuroprotection and neuroregeneration against Aβ toxicity, as well as to cross the BBB offering a potential lead for AD therapeutics.
Collapse
Affiliation(s)
- Krishnangsu Pradhan
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, 700032 West Bengal, India
| | - Gaurav Das
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, 700032 West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology Campus, 4 Raja S. C. Mullick Road, Kolkata 700032, India
| | - Varsha Gupta
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, 700032 West Bengal, India
| | - Prasenjit Mondal
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, 700032 West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology Campus, 4 Raja S. C. Mullick Road, Kolkata 700032, India
| | - Surajit Barman
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, 700032 West Bengal, India
| | - Juhee Khan
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, 700032 West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology Campus, 4 Raja S. C. Mullick Road, Kolkata 700032, India
| | - Surajit Ghosh
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, 700032 West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology Campus, 4 Raja S. C. Mullick Road, Kolkata 700032, India
| |
Collapse
|
33
|
Xu J, Yuan Y, Zhang R, Song Y, Sui T, Wang J, Wang C, Chen Y, Guan S, Wang L. A deuterohemin peptide protects a transgenic Caenorhabditis elegans model of Alzheimer’s disease by inhibiting Aβ1–42 aggregation. Bioorg Chem 2019; 82:332-339. [DOI: 10.1016/j.bioorg.2018.10.072] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/27/2018] [Accepted: 10/31/2018] [Indexed: 01/05/2023]
|
34
|
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: 16] [Impact Index Per Article: 2.7] [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
|
35
|
Pradhan K, Das G, Mondal P, Khan J, Barman S, Ghosh S. Genesis of Neuroprotective Peptoid from Aβ30-34 Inhibits Aβ Aggregation and AChE Activity. ACS Chem Neurosci 2018; 9:2929-2940. [PMID: 30036464 DOI: 10.1021/acschemneuro.8b00071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Aβ peptide and hyper-phosphorylated microtubule associated protein (Tau) aggregation causes severe damage to both the neuron membrane and key signal processing microfilament (microtubule) in Alzheimer's disease (AD) brains. To date, the key challenge is to develop nontoxic, proteolytically stable amyloid inhibitors, which can simultaneously target multiple pathways involved in AD. Various attempts have been made in this direction; however, clinical outcomes of those attempts have been reported to be poor. Thus, we choose development of peptoid (N-substituted glycine oligomers)-based leads as potential AD therapeutics, which are easy to synthesize, found to be proteolytically stable, and exhibit excellent bioavailability. In this paper, we have designed and synthesized a new short peptoid for amyloid inhibition from 30-34 hydrophobic pocket of amyloid beta (Aβ) peptide. The peptoid selectively binds with 17-21 hydrophobic region of Aβ and inhibits Aβ fibril formation. Various in vitro assays suggested that our AI peptoid binds with tubulin/microtubule and promotes its polymerization and stability. This peptoid also inhibits AChE-induced Aβ fibril formation and provides significant neuroprotection against toxicity generated by nerve growth factor (NGF) deprived neurons derived from rat adrenal pheochromocytoma (PC12) cell line. Moreover, this peptoid shows serum stability and is noncytotoxic to primary rat cortical neurons.
Collapse
Affiliation(s)
- Krishnangsu Pradhan
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India
| | - Gaurav Das
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology Campus, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Prasenjit Mondal
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology Campus, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Juhee Khan
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology Campus, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Surajit Barman
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India
| | - Surajit Ghosh
- Organic & Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Chemical Biology Campus, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| |
Collapse
|
36
|
Zhang X, Fu Z, Meng L, He M, Zhang Z. The Early Events That Initiate β-Amyloid Aggregation in Alzheimer's Disease. Front Aging Neurosci 2018; 10:359. [PMID: 30542277 PMCID: PMC6277872 DOI: 10.3389/fnagi.2018.00359] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 10/22/2018] [Indexed: 12/18/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by the development of amyloid plaques and neurofibrillary tangles (NFTs) consisting of aggregated β-amyloid (Aβ) and tau, respectively. The amyloid hypothesis has been the predominant framework for research in AD for over two decades. According to this hypothesis, the accumulation of Aβ in the brain is the primary factor initiating the pathogenesis of AD. However, it remains elusive what factors initiate Aβ aggregation. Studies demonstrate that AD has multiple causes, including genetic and environmental factors. Furthermore, genetic factors, many age-related events and pathological conditions such as diabetes, traumatic brain injury (TBI) and aberrant microbiota also affect the aggregation of Aβ. Here we provide an overview of the age-related early events and other pathological processes that precede Aβ aggregation.
Collapse
Affiliation(s)
- Xingyu Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhihui Fu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lanxia Meng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mingyang He
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
37
|
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: 1.1] [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
|
38
|
Herline K, Prelli F, Mehta P, MacMurray C, Goñi F, Wisniewski T. Immunotherapy to improve cognition and reduce pathological species in an Alzheimer's disease mouse model. ALZHEIMERS RESEARCH & THERAPY 2018; 10:54. [PMID: 29914551 PMCID: PMC6006698 DOI: 10.1186/s13195-018-0384-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/10/2018] [Indexed: 12/11/2022]
Abstract
Background Alzheimer’s disease (AD) is characterized by physiologically endogenous proteins amyloid beta (Aβ) and tau undergoing a conformational change and accumulating as soluble oligomers and insoluble aggregates. Tau and Aβ soluble oligomers, which contain extensive β-sheet secondary structure, are thought to be the most toxic forms. The objective of this study was to determine the ability of TWF9, an anti-β-sheet conformation antibody (aβComAb), to selectively recognize pathological Aβ and phosphorylated tau in AD human tissue compared with cognitively normal age-matched controls and to improve the performance of old 3xTg-AD mice with advanced pathology in behavioral testing after acute treatment with TWF9. Methods In this study, we used immunohistochemistry, immunoprecipitation, and enzyme-linked immunosorbent assay (ELISA) to characterize TWF9 specificity. We further assessed cognitive performance in old (18–22 months) 3xTg-AD mice using both a Barnes maze and novel object recognition after intraperitoneal administration of TWF9 (4 mg/kg) biweekly for 2 weeks before the start of behavioral testing. Injections continued for the duration of the behavioral testing, which lasted 2 weeks. Results Histological analysis of TWF9 in formalin-fixed paraffin-embedded human control and AD (ABC score: A3B3C3) brain tissue revealed preferential cytoplasmic immunoreactivity in neurons in the AD tissue compared with controls (p < 0.05). Furthermore, ELISA using oligomeric and monomeric Aβ showed a preferential affinity for oligomeric Aβ. Immunoprecipitation studies showed that TWF9 extracted both phosphorylated tau (p < 0.01) and Aβ (p < 0.01) from fresh frozen brain tissues. Results show that treated old 3xTg-AD mice have an enhanced novel object recognition memory (p < 0.01) and Barnes maze performance (p = 0.05) compared with control animals. Overall plaque burden, neurofibrillary tangles, microgliosis, and astrocytosis remained unchanged. Soluble phosphorylated tau was significantly reduced in TWF9-treated mice (p < 0.05), and there was a trend for a reduction in soluble Aβ levels in the brain homogenates of female 3xTg-AD mice (p = 0.06). Conclusions This study shows that acute treatment with an aβComAb can effectively improve performance in behavioral testing without reduction of amyloid plaque burden, and that peripherally administered IgG can affect levels of pathological species in the brain.
Collapse
Affiliation(s)
- Krystal Herline
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA
| | - Frances Prelli
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA
| | - Pankaj Mehta
- Department of Immunology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, USA
| | | | - Fernando Goñi
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA
| | - Thomas Wisniewski
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA. .,Departments of Pathology and Psychiatry, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
39
|
Hu Z, Yang Y, Zhao Y, Yu H, Ying X, Zhou D, Zhong J, Zheng Z, Liu J, Pan R, Zhang W, Cheng F, Duan S. APOE hypermethylation is associated with autism spectrum disorder in a Chinese population. Exp Ther Med 2018; 15:4749-4754. [PMID: 29844799 PMCID: PMC5958870 DOI: 10.3892/etm.2018.6069] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 03/01/2018] [Indexed: 12/27/2022] Open
Abstract
Abnormal apolipoprotein E (APOE) methylation has been demonstrated to be associated with Alzheimer's disease, which may have overlapping mechanisms with autism spectrum disorder (ASD). Thus, the purpose of the present study was to assess the possible link between APOE methylation and ASD. Genomic DNA was extracted from peripheral blood and subjected to a methylation assay. SYBR green-based quantitative methylation-specific polymerase chain reaction analysis was used to measure APOE methylation in 62 pediatric patients with ASD and 73 age-matched healthy subjects. The APOE methylation in each sample was expressed as a percentage of methylation of a reference (PMR). The results indicated that APOE methylation in pediatric patients with ASD was significantly higher than that in the healthy controls (median PMR, 33 vs. 11%; P=2.36×10−10). Receiver operating characteristic curve demonstrated that PMR of 15.4% was the optimal cut-off for predicting ASD (area under curve, 0.817; sensitivity, 93.5%; specificity, 72.6%; P=2.36×10−10). In summary, the present results indicated that APOE hypermethylation in peripheral blood DNA may be used as a diagnostic biomarker for ASD.
Collapse
Affiliation(s)
- Zhenyu Hu
- Department of Child Psychiatry, Ningbo Kangning Hospital, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Yong Yang
- Zhejiang Provincial Key Laboratory of Pathophysiology, Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Yuanzhi Zhao
- Department of Child Psychiatry, Ningbo Kangning Hospital, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Hang Yu
- Zhejiang Provincial Key Laboratory of Pathophysiology, Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Xiuru Ying
- Zhejiang Provincial Key Laboratory of Pathophysiology, Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Dongsheng Zhou
- Department of Child Psychiatry, Ningbo Kangning Hospital, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Jie Zhong
- Zhejiang Provincial Key Laboratory of Pathophysiology, Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Zhonghua Zheng
- Zhejiang Provincial Key Laboratory of Pathophysiology, Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Jing Liu
- Zhejiang Provincial Key Laboratory of Pathophysiology, Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Ranran Pan
- Zhejiang Provincial Key Laboratory of Pathophysiology, Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Wenwu Zhang
- Department of Child Psychiatry, Ningbo Kangning Hospital, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Fang Cheng
- Department of Child Psychiatry, Ningbo Kangning Hospital, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| | - Shiwei Duan
- Zhejiang Provincial Key Laboratory of Pathophysiology, Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
40
|
Young SC. A Systematic Review of Antiamyloidogenic and Metal-Chelating Peptoids: Two Structural Motifs for the Treatment of Alzheimer's Disease. Molecules 2018; 23:E296. [PMID: 29385058 PMCID: PMC6017092 DOI: 10.3390/molecules23020296] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 01/27/2018] [Accepted: 01/30/2018] [Indexed: 11/17/2022] Open
Abstract
Alzheimer's disease (AD) is an incurable form of dementia affecting millions of people worldwide and costing billions of dollars in health care-related payments, making the discovery of a cure a top health, societal, and economic priority. Peptide-based drugs and immunotherapies targeting AD-associated beta-amyloid (Aβ) aggregation have been extensively explored; however, their therapeutic potential is limited by unfavorable pharmacokinetic (PK) properties. Peptoids (N-substituted glycine oligomers) are a promising class of peptidomimetics with highly tunable secondary structures and enhanced stabilities and membrane permeabilities. In this review, the biological activities, structures, and physicochemical properties for several amyloid-targeting peptoids will be described. In addition, metal-chelating peptoids with the potential to treat AD will be discussed since there are connections between the dysregulation of certain metals and the amyloid pathway.
Collapse
Affiliation(s)
- Sherri C Young
- Department of Chemistry, Muhlenberg College, 2400 Chew Street, Allentown, PA 18104, USA.
| |
Collapse
|
41
|
Goñi F, Martá-Ariza M, Herline K, Peyser D, Boutajangout A, Mehta P, Drummond E, Prelli F, Wisniewski T. Anti-β-sheet conformation monoclonal antibody reduces tau and Aβ oligomer pathology in an Alzheimer's disease model. ALZHEIMERS RESEARCH & THERAPY 2018; 10:10. [PMID: 29378642 PMCID: PMC5789573 DOI: 10.1186/s13195-018-0337-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 01/04/2018] [Indexed: 02/08/2023]
Abstract
Background Oligomeric forms of amyloid-β (Aβ) and tau are increasing being recognized as key toxins in the pathogenesis of Alzheimer’s disease (AD). Methods We developed a novel monoclonal antibody (mAb), GW-23B7, that recognizes β-sheet secondary structure on pathological oligomers of neurodegenerative diseases. Results The pentameric immunoglobulin M kappa chain (IgMκp) we developed specifically distinguishes intra- and extracellular pathology in human AD brains. Purified GW-23B7 showed a dissociation constant in the nanomolar range for oligomeric Aβ and did not bind monomeric Aβ. In enzyme-linked immunosorbent assays, it recognized oligomeric forms of both Aβ and hyperphosphorylated tau. Aged triple-transgenic AD mice with both Aβ and tau pathology infused intraperitoneally for 2 months showed IgMκp in the soluble brain homogenate, peaking at 24 h postinoculation. Treated mice exhibited significant cognitive rescue on radial arm maze testing compared with vehicle control-infused mice. Immunohistochemically, treatment resulted in a significant decrease of extracellular pathology. Biochemically, treatment resulted in significant reductions of oligomeric forms of Aβ and tau. Conclusions These results suggest that GW-23B7, an anti-β-sheet conformational mAb humanized for clinical trials, may be an effective therapeutic agent for human AD. Electronic supplementary material The online version of this article (10.1186/s13195-018-0337-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fernando Goñi
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA.
| | - Mitchell Martá-Ariza
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA
| | - Krystal Herline
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA
| | - Daniel Peyser
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA
| | - Allal Boutajangout
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA.,Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Pankaj Mehta
- Department of Immunology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Eleanor Drummond
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA
| | - Frances Prelli
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA
| | - Thomas Wisniewski
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA. .,Department of Pathology, New York University School of Medicine, New York, NY, USA. .,Department of Psychiatry, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|