1
|
Cantero-Fortiz Y, Boada M. The use of plasma exchange with albumin replacement in the management of Alzheimer's disease: a scoping review. Front Neurol 2024; 15:1443132. [PMID: 39421573 PMCID: PMC11484623 DOI: 10.3389/fneur.2024.1443132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/26/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction AD is a progressive neurodegenerative disorder causing significant cognitive decline and impaired daily functioning. Current treatments offer only modest relief, and many amyloid-targeting therapies have failed, prompting exploration of alternative approaches such as PE with albumin replacement. Objectives This scoping review systematically maps the literature on PE with albumin replacement in AD management, focusing on outcomes, methodologies, and reported benefits and risks. Methods A comprehensive search in PubMed, supplemented by reference scanning and hand-searching, identified studies involving PE with albumin replacement in AD patients. Data charting and critical appraisal were conducted using standardized tools. Results Seven primary studies from the AMBAR (Alzheimer Management by Albumin Replacement) trial met the inclusion criteria, consistently reporting improvements in cognitive function, positive neuroimaging results, and favorable neuropsychiatric outcomes. For instance, one study found a significant slowing of cognitive decline (p < 0.05) among patients receiving PE with albumin replacement. Another study showed better preservation of hippocampal volume and improved brain perfusion metrics in the treatment group (p < 0.05). The intervention was generally well-tolerated with manageable side effects. Conclusion PE with albumin replacement is a promising therapeutic approach for AD, warranting further investigation to confirm its efficacy and safety across broader settings. Scoping review registration https://osf.io/v6dez/?view_only=1cd9637e7e0347d39713bf19aac0dfe8.
Collapse
Affiliation(s)
- Yahveth Cantero-Fortiz
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
2
|
Aranda-Abreu GE, Rojas-Durán F, Hernández-Aguilar ME, Herrera-Covarrubias D, Chí-Castañeda LD, Toledo-Cárdenas MR, Suárez-Medellín JM. Alzheimer's Disease: Cellular and Pharmacological Aspects. Geriatrics (Basel) 2024; 9:86. [PMID: 39051250 PMCID: PMC11270425 DOI: 10.3390/geriatrics9040086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/23/2024] [Accepted: 06/21/2024] [Indexed: 07/27/2024] Open
Abstract
Alzheimer's disease was described more than 100 years ago and despite the fact that several molecules are being tested for its treatment, which are in phase III trials, the disease continues to progress. The main problem is that these molecules function properly in healthy neurons, while neuronal pathology includes plasma membrane disruption, malfunction of various organelles, and hyperphosphorylation of Tau and amyloid plaques. The main objective of this article is the discussion of a neuronal restoration therapy, where molecules designed for the treatment of Alzheimer's disease would probably be more effective, and the quality of life of people would be better.
Collapse
Affiliation(s)
- Gonzalo Emiliano Aranda-Abreu
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa 91192, Mexico; (F.R.-D.); (M.E.H.-A.); (D.H.-C.); (L.D.C.-C.); (M.R.T.-C.); (J.M.S.-M.)
| | | | | | | | | | | | | |
Collapse
|
3
|
Blomgren F, Rodin A, Chrobak W, Pacut DW, Swenson J, Ermilova I. Two statins and cromolyn as possible drugs against the cytotoxicity of Aβ(31-35) and Aβ(25-35) peptides: a comparative study by advanced computer simulation methods. RSC Adv 2022; 12:13352-13366. [PMID: 35520132 PMCID: PMC9066867 DOI: 10.1039/d2ra01963a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 04/19/2022] [Indexed: 11/21/2022] Open
Abstract
In this work, possible effective mechanisms of cromolyn, atorvastatin and lovastatin on the cytotoxicity of Aβ(31-35) and Aβ(25-35) peptides were investigated by classical molecular dynamics and well-tempered metadynamics simulations. The results demonstrate that all the drugs affect the behavior of the peptides, such as their ability to aggregate, and alter their secondary structures and their affinity to a particular drug. Our findings from the computed properties suggest that the best drug candidate is lovastatin. This medicine inhibits peptide aggregation, adsorbs the peptides on the surface of the drug clusters, changes the secondary structure and binds to MET35, which has been seen as the reason for the toxicity of the studied peptide sequences. Moreover, lovastatin is the drug which previously has demonstrated the strongest ability to penetrate the blood-brain barrier and makes lovastatin the most promising medicine among the three investigated drugs. Atorvastatin is also seen as a potential candidate if its penetration through the blood-brain barrier could be improved. Otherwise, its properties are even better than the ones demonstrated by lovastatin. Cromolyn appears to be less interesting as an anti-aggregant from the computational data, in comparison to the two statins.
Collapse
Affiliation(s)
- Fredrik Blomgren
- Department of Physics, Chalmers University of Technology Fysikgränd 4 Göteborg 41258 Sweden +46-728487773
| | - Alexander Rodin
- Department of Physics, Chalmers University of Technology Fysikgränd 4 Göteborg 41258 Sweden +46-728487773
| | - Wojciech Chrobak
- Department of Physics, Chalmers University of Technology Fysikgränd 4 Göteborg 41258 Sweden +46-728487773
| | - Dawid Wojciech Pacut
- Department of Physics, Chalmers University of Technology Fysikgränd 4 Göteborg 41258 Sweden +46-728487773
| | - Jan Swenson
- Department of Physics, Chalmers University of Technology Fysikgränd 4 Göteborg 41258 Sweden +46-728487773
| | - Inna Ermilova
- Department of Physics, Chalmers University of Technology Fysikgränd 4 Göteborg 41258 Sweden +46-728487773
| |
Collapse
|
4
|
Wang X, Sun H, Pan S, Bai X, Zhu Z, Zhang R, Li C, Chen Y, Bao M, Zhang K, Feng R. Causal Relationships Between Relative Intake from the Macronutrients and Alzheimer's Disease: A Two-Sample Mendelian Randomization Study. J Alzheimers Dis 2022; 87:665-673. [PMID: 35342086 DOI: 10.3233/jad-215535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Some observational studies indicated the associations of relative carbohydrate, sugar, fat, and protein intake and Alzheimer's disease (AD). But it remains unclear whether the associations are causal. OBJECTIVE This study aimed to identify the effects of relative carbohydrate, sugar, fat, and protein intake in the diet on AD. METHODS A two-sample Mendelian randomization was employed. Finally, 14 independent lead SNPs remained in the Social Science Genetic Association Consortium. These SNPs of relative carbohydrate, sugar, fat, and protein intake at the level of genome-wide significance (p < 5×10-8) were used as instrumental variables. The summary data for AD were acquired from the International Genomics of Alzheimer's Project with a total of 54,162 individuals (17,008 AD patients and 37,154 control participants). RESULTS This two-sample Mendelian randomization indicated that increased relative protein intake (per 1 standard deviation) causally decreased the AD risk (OR = 0.48, 95% CI: 0.24-0.95, p = 0.036), and increased relative fat intake may decrease the risk of AD (OR = 0.22, 95% CI: 0.06-0.86, p = 0.029). No statistical significance with AD risk was seen for relative carbohydrate or relative sugar intake. CONCLUSION A higher relative intake of protein can causally reduce the risk of AD in the elderly. Additionally, a higher relative intake of fat may be protective against AD. No evidence showed that AD was associated with relative carbohydrate and sugar intake.
Collapse
Affiliation(s)
- Xiaoxin Wang
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Hongru Sun
- Department of Epidemiology, School of Public Health, Harbin Medical University, Harbin, Heilongjiang Province, P. R. China
| | - Sijia Pan
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Xiao Bai
- Haxi New Area Community Health Service Center, Nangang District, Harbin, Heilongjiang Province, China
| | - Zhuolin Zhu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Runan Zhang
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Chunlong Li
- Department of General Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Chen
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Meitong Bao
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Kewei Zhang
- Department of Mathematics, Heilongjiang Institute of Technology, Harbin, China
| | - Rennan Feng
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| |
Collapse
|
5
|
Beshir SA, Aadithsoorya AM, Parveen A, Goh SSL, Hussain N, Menon VB. Aducanumab Therapy to Treat Alzheimer's Disease: A Narrative Review. Int J Alzheimers Dis 2022; 2022:9343514. [PMID: 35308835 PMCID: PMC8926483 DOI: 10.1155/2022/9343514] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 02/16/2022] [Indexed: 12/19/2022] Open
Abstract
Background Aducanumab, a new monoclonal antibody that targets β-amyloid aggregates, has been granted conditional approval by the U.S. FDA for treatment of mild Alzheimer's disease (AD). The approval of this drug without a confirmed significant clinical impact has resulted in several debates. Objective In this narrative review, aducanumab approval-related controversy, the drug's pharmacokinetics and pharmacodynamic characteristics, evidence from the efficacy and safety trials of aducanumab, implications of the drug approval, and the future directions in the management of patients with AD are summarized. Methods Using relevant keywords, Google Scholar, Web of Science, and MEDLINE databases and manufacturer's website were searched. Results Infusion of aducanumab at a higher dose resulted in a modest slowing of cognitive decline among patients with mild cognitive impairment or early-onset AD dementia. The drug however can cause amyloid-related imaging abnormalities. Due to modest impact on cognition, the use of this drug by patients with AD will most likely be limited. The manufacturer is required to run an extended phase IIIb trial to verify the benefit of this drug. Access to therapy requires a careful selection of patients and periodic monitoring to ensure the optimal use of the drug. Conclusion Despite the limitations, aducanumab is the first disease-modifying therapy approved for the treatment of AD. Aducanumab addresses a part of the pathogenesis of AD; therefore, drugs that can act on multiple targets are needed. In addition, the search for preventive strategies, validated plasma-based assays, and newer drugs for AD, which are effective, safe, convenient, and affordable, is vital.
Collapse
Affiliation(s)
- Semira Abdi Beshir
- Clinical Pharmacy & Pharmacotherapeutics Department, Dubai Pharmacy College, Dubai, UAE
| | | | - Affana Parveen
- College of Pharmacy, Gulf Medical University, Ajman, UAE
| | - Sheron Sir Loon Goh
- Department of Primary Care Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Nadia Hussain
- Department of Pharmaceutical Sciences, College of Pharmacy, Al Ain University, Al Ain, Abu Dhabi, UAE
| | | |
Collapse
|
6
|
Gutierrez L, Folch A, Rojas M, Cantero JL, Atienza M, Folch J, Camins A, Ruiz A, Papandreou C, Bulló M. Effects of Nutrition on Cognitive Function in Adults with or without Cognitive Impairment: A Systematic Review of Randomized Controlled Clinical Trials. Nutrients 2021; 13:nu13113728. [PMID: 34835984 PMCID: PMC8621754 DOI: 10.3390/nu13113728] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/13/2021] [Accepted: 10/19/2021] [Indexed: 01/06/2023] Open
Abstract
New dietary approaches for the prevention of cognitive impairment are being investigated. However, evidence from dietary interventions is mainly from food and nutrient supplement interventions, with inconsistent results and high heterogeneity between trials. We conducted a comprehensive systematic search of randomized controlled trials (RCTs) published in MEDLINE-PubMed, from January 2018 to July 2021, investigating the impact of dietary counseling, as well as food-based and dietary supplement interventions on cognitive function in adults with or without cognitive impairment. Based on the search strategy, 197 eligible publications were used for data abstraction. Finally, 61 articles were included in the analysis. There was reasonable evidence that dietary patterns, as well as food and dietary supplements improved cognitive domains or measures of brain integrity. The Mediterranean diet showed promising results, whereas the role of the DASH diet was not clear. Healthy food consumption improved cognitive function, although the quality of these studies was relatively low. The role of dietary supplements was mixed, with strong evidence of the benefits of polyphenols and combinations of nutrients, but with low evidence for PUFAs, vitamin D, specific protein, amino acids, and other types of supplements. Further well-designed RCTs are needed to guide the development of dietary approaches for the prevention of cognitive impairment.
Collapse
Affiliation(s)
- Laia Gutierrez
- Nutrition and Metabolic Disorders Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University, 43201 Reus, Spain; (L.G.); (A.F.); (M.R.); (J.F.)
- Nutrition and Metabolic Disorders Research Group, Institute of Health Pere Virgili—IISPV, 43204 Reus, Spain;
| | - Alexandre Folch
- Nutrition and Metabolic Disorders Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University, 43201 Reus, Spain; (L.G.); (A.F.); (M.R.); (J.F.)
- Nutrition and Metabolic Disorders Research Group, Institute of Health Pere Virgili—IISPV, 43204 Reus, Spain;
| | - Melina Rojas
- Nutrition and Metabolic Disorders Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University, 43201 Reus, Spain; (L.G.); (A.F.); (M.R.); (J.F.)
- Nutrition and Metabolic Disorders Research Group, Institute of Health Pere Virgili—IISPV, 43204 Reus, Spain;
| | - José Luis Cantero
- Laboratory of Functional Neuroscience, Pablo de Olavide University, 41013 Seville, Spain; (J.L.C.); (M.A.)
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain; (A.C.); (A.R.)
| | - Mercedes Atienza
- Laboratory of Functional Neuroscience, Pablo de Olavide University, 41013 Seville, Spain; (J.L.C.); (M.A.)
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain; (A.C.); (A.R.)
| | - Jaume Folch
- Nutrition and Metabolic Disorders Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University, 43201 Reus, Spain; (L.G.); (A.F.); (M.R.); (J.F.)
- Nutrition and Metabolic Disorders Research Group, Institute of Health Pere Virgili—IISPV, 43204 Reus, Spain;
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain; (A.C.); (A.R.)
| | - Antoni Camins
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain; (A.C.); (A.R.)
- Department of Pharmacology, Toxicology & Therapeutic Chemistry, Faculty of Pharmacy & Food Sciences, University of Barcelona, 08028 Barcelona, Spain
- Institut de Neurociències (UBNeuro), University of Barcelona, 08035 Barcelona, Spain
| | - Agustín Ruiz
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, 28031 Madrid, Spain; (A.C.); (A.R.)
- ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), 08028 Barcelona, Spain
| | - Christopher Papandreou
- Nutrition and Metabolic Disorders Research Group, Institute of Health Pere Virgili—IISPV, 43204 Reus, Spain;
- CIBER Physiology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, 28029 Madrid, Spain
| | - Mònica Bulló
- Nutrition and Metabolic Disorders Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University, 43201 Reus, Spain; (L.G.); (A.F.); (M.R.); (J.F.)
- Nutrition and Metabolic Disorders Research Group, Institute of Health Pere Virgili—IISPV, 43204 Reus, Spain;
- CIBER Physiology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-97-775-9388
| |
Collapse
|
7
|
Król S, Österlund N, Vosough F, Jarvet J, Wärmländer S, Barth A, Ilag LL, Magzoub M, Gräslund A, Mörman C. The amyloid-inhibiting NCAM-PrP peptide targets Aβ peptide aggregation in membrane-mimetic environments. iScience 2021; 24:102852. [PMID: 34381976 PMCID: PMC8340127 DOI: 10.1016/j.isci.2021.102852] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/27/2021] [Accepted: 07/09/2021] [Indexed: 01/16/2023] Open
Abstract
Substantial research efforts have gone into elucidating the role of protein misfolding and self-assembly in the onset and progression of Alzheimer's disease (AD). Aggregation of the Amyloid-β (Aβ) peptide into insoluble fibrils is closely associated with AD. Here, we use biophysical techniques to study a peptide-based approach to target Aβ amyloid aggregation. A peptide construct, NCAM-PrP, consists of a largely hydrophobic signal sequence linked to a positively charged hexapeptide. The NCAM-PrP peptide inhibits Aβ amyloid formation by forming aggregates which are unavailable for further amyloid aggregation. In a membrane-mimetic environment, Aβ and NCAM-PrP form specific heterooligomeric complexes, which are of lower aggregation states compared to Aβ homooligomers. The Aβ:NCAM-PrP interaction appears to take place on different aggregation states depending on the absence or presence of a membrane-mimicking environment. These insights can be useful for the development of potential future therapeutic strategies targeting Aβ at several aggregation states.
Collapse
Affiliation(s)
- Sylwia Król
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, 106 91, Sweden
| | - Nicklas Österlund
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, 106 91, Sweden
| | - Faraz Vosough
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, 106 91, Sweden
| | - Jüri Jarvet
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, 106 91, Sweden
| | - Sebastian Wärmländer
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, 106 91, Sweden
| | - Andreas Barth
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, 106 91, Sweden
| | - Leopold L. Ilag
- Department of Materials and Environmental Chemistry, Stockholm University, Stockholm, 106 91, Sweden
| | - Mazin Magzoub
- Biology Program, Division of Science, New York University Abu Dhabi, Box 129188, Abu Dhabi, United Arab Emirates
| | - Astrid Gräslund
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, 106 91, Sweden
| | - Cecilia Mörman
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, 106 91, Sweden
| |
Collapse
|
8
|
Angehrn Z, Sostar J, Nordon C, Turner A, Gove D, Karcher H, Keenan A, Mittelstadt B, de Reydet-de Vulpillieres F. Ethical and Social Implications of Using Predictive Modeling for Alzheimer's Disease Prevention: A Systematic Literature Review. J Alzheimers Dis 2021; 76:923-940. [PMID: 32597799 DOI: 10.3233/jad-191159] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The therapeutic paradigm in Alzheimer's disease (AD) is shifting from symptoms management toward prevention goals. Secondary prevention requires the identification of individuals without clinical symptoms, yet "at-risk" of developing AD dementia in the future, and thus, the use of predictive modeling. OBJECTIVE The objective of this study was to review the ethical concerns and social implications generated by this new approach. METHODS We conducted a systematic literature review in Medline, Embase, PsycInfo, and Scopus, and complemented it with a gray literature search between March and July 2018. Then we analyzed data qualitatively using a thematic analysis technique. RESULTS We identified thirty-one ethical issues and social concerns corresponding to eight ethical principles: (i) respect for autonomy, (ii) beneficence, (iii) non-maleficence, (iv) equality, justice, and diversity, (v) identity and stigma, (vi) privacy, (vii) accountability, transparency, and professionalism, and (viii) uncertainty avoidance. Much of the literature sees the discovery of disease-modifying treatment as a necessary and sufficient condition to justify AD risk assessment, overlooking future challenges in providing equitable access to it, establishing long-term treatment outcomes and social consequences of this approach, e.g., medicalization. The ethical/social issues associated specifically with predictive models, such as the adequate predictive power and reliability, infrastructural requirements, data privacy, potential for personalized medicine in AD, and limiting access to future AD treatment based on risk stratification, were covered scarcely. CONCLUSION The ethical discussion needs to advance to reflect recent scientific developments and guide clinical practice now and in the future, so that necessary safeguards are implemented for large-scale AD secondary prevention.
Collapse
|
9
|
Qiao O, Ji H, Zhang Y, Zhang X, Zhang X, Liu N, Huang L, Liu C, Gao W. New insights in drug development for Alzheimer's disease based on microglia function. Biomed Pharmacother 2021; 140:111703. [PMID: 34083109 DOI: 10.1016/j.biopha.2021.111703] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 12/26/2022] Open
Abstract
One of the biggest challenges in drug development for Alzheimer's disease (AD) is how to effectively remove deposits of amyloid-beta (Aβ). Recently, the relationship between microglia and Aβ has become a research hotspot. Emerging evidence suggests that Aβ-induced microglia-mediated neuroinflammation further aggravates the decline of cognitive function, while microglia are also involved in the process of Aβ clearance. Hence, microglia have become a potential therapeutic target for the treatment or prevention of AD. An in-depth understanding of the role played by microglia in the development of AD will help us to broaden therapeutic strategies for AD. In this review, we provide an overview of the dual roles of microglia in AD progression: the positive effect of phagocytosis of Aβ and its negative effect on neuroinflammation after over-activation. With the advantages of novel structure, high efficiency, and low toxicity, small-molecule compounds as modulators of microglial function have attracted considerable attention in the therapeutic areas of AD. In this review, we also summarize the therapeutic potential of small molecule compounds (SMCs) and their structure-activity relationship for AD treatment through modulating microglial phagocytosis and inhibiting neuroinflammation. For example, the position and number of phenolic hydroxyl groups on the B ring are the key to the activity of flavonoids, and the substitution of hydroxyl groups on the benzene ring enhances the anti-inflammatory activity of phenolic acids. This review is expected to be useful for developing effective modulators of microglial function from SMCs for the amelioration and treatment of AD.
Collapse
Affiliation(s)
- Ou Qiao
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China
| | - Haixia Ji
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China
| | - Yi Zhang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China
| | - Xinyu Zhang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China
| | - Xueqian Zhang
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China
| | - Na Liu
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China
| | - Luqi Huang
- Chinese Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Changxiao Liu
- The State Key Laboratories of Pharmacodynamics and Pharmacokinetics, Tianjin 300193, China
| | - Wenyuan Gao
- Tianjin Key Laboratory for Modern Drug Delivery and High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Weijin Road, Tianjin 300072, China.
| |
Collapse
|
10
|
Kowalska M, Wize K, Prendecki M, Lianeri M, Kozubski W, Dorszewska J. Genetic Variants and Oxidative Stress in Alzheimer's Disease. Curr Alzheimer Res 2021; 17:208-223. [PMID: 32091332 DOI: 10.2174/1567205017666200224121447] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 01/08/2020] [Accepted: 02/12/2020] [Indexed: 12/26/2022]
Abstract
In an aging society, the number of people suffering from Alzheimer's Disease (AD) is still growing. Currently, intensive research is being carried out on the pathogenesis of AD. The results of these studies indicated that oxidative stress plays an important role in the onset and development of this disease. Moreover, in AD oxidative stress is generated by both genetic and biochemical factors as well as the functioning of the systems responsible for their formation and removal. The genetic factors associated with the regulation of the redox system include TOMM40, APOE, LPR, MAPT, APP, PSEN1 and PSEN2 genes. The most important biochemical parameters related to the formation of oxidative species in AD are p53, Homocysteine (Hcy) and a number of others. The formation of Reactive Oxygen Species (ROS) is also related to the efficiency of the DNA repair system, the effectiveness of the apoptosis, autophagy and mitophagy processes as well as the antioxidant potential. However, these factors are responsible for the development of many disorders, often with similar clinical symptoms, especially in the early stages of the disease. The discovery of markers of the early diagnosis of AD may contribute to the introduction of pharmacotherapy and slow down the progression of this disease.
Collapse
Affiliation(s)
- Marta Kowalska
- Laboratory of Neurobiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Wize
- Laboratory of Neurobiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Michał Prendecki
- Laboratory of Neurobiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Margarita Lianeri
- Laboratory of Neurobiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Wojciech Kozubski
- Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | - Jolanta Dorszewska
- Laboratory of Neurobiology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
11
|
Page TH, Chiappo D, Brunini F, Garnica J, Blackburn J, Dudhiya F, Prendecki M, McAdoo SP, Pusey CD. Danger-associated molecular pattern molecules and the receptor for advanced glycation end products enhance ANCA-induced responses. Rheumatology (Oxford) 2021; 61:834-845. [PMID: 33974049 PMCID: PMC8824420 DOI: 10.1093/rheumatology/keab413] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 05/04/2021] [Indexed: 11/25/2022] Open
Abstract
Objectives The pro-inflammatory activities of the calgranulins and HMGB1 can be counteracted by sRAGE, the soluble form of their shared receptor. To understand the role of these molecules in AAV and their potential as therapeutic targets we have studied (i) the relationship between these DAMPS and disease activity; (ii) the expression of RAGE and sRAGE in biopsy tissue and peripheral blood; and (iii) the effect of these molecules on ANCA-mediated cytokine production. Methods We examined circulating levels of calgranulins (S100A8/A9 and S100A12), HMGB1 and sRAGE by ELISA. RAGE was examined in AAV kidney and lung biopsies by immunohistochemistry and RAGE expression was monitored in peripheral blood by qPCR. In vitro, the effect of co-stimulating PBMC with ANCA and S100A8/A9 on cytokine production was studied by ELISA. Results We found significantly raised levels of calgranulins and HMGB1 in active AAV regardless of clinical phenotype (PR3+/MPO+ AAV). Levels of calgranulins showed significant correlations with each other. RAGE protein and message was raised in peripheral blood and in cells infiltrating kidney and lung biopsy tissue, while sRAGE was lowered. Furthermore, ANCA-mediated production of IL-8 from PBMC was significantly enhanced by the presence of S100A8/A9 in a RAGE/TLR4-dependent manner. Conclusions Raised circulating calgranulins provide a good marker of disease activity in AAV and are unlikely to be counteracted by sRAGE. Increased RAGE expression in AAV indicates receptor stimulation in active disease that may exacerbate ANCA-induced cytokine production. Targeting the RAGE pathway may provide a useful therapeutic approach in AAV.
Collapse
Affiliation(s)
- Theresa H Page
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, UK
| | - Derick Chiappo
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, UK
| | - Francesca Brunini
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, UK.,Nephrology and Dialysis Unit, Ospedale di Circolo e Fondazione Macchi, ASST-Settelaghi, Varese, Italy
| | - Josep Garnica
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, UK.,Institut D'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, 08036, Spain
| | - Jack Blackburn
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, UK
| | - Fayaz Dudhiya
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, UK
| | - Maria Prendecki
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, UK
| | - Stephen P McAdoo
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, UK
| | - Charles D Pusey
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
12
|
Liori S, Arfaras-Melainis A, Bistola V, Polyzogopoulou E, Parissis J. Cognitive impairment in heart failure: clinical implications, tools of assessment, and therapeutic considerations. Heart Fail Rev 2021; 27:993-999. [PMID: 33939080 DOI: 10.1007/s10741-021-10118-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/26/2021] [Indexed: 10/21/2022]
Abstract
Cognitive impairment (CI) is an important comorbidity in patients with heart failure (HF). Its prevalence parallels the severity of heart failure, while it is an independent prognostic marker of adverse events. Various factors contribute to cognitive decline in HF, influencing self-care. There are no standardized screening methods for the diagnosis and management of these patients. The aim of the present manuscript is to provide an overview of the impact of cognitive impairment in HF, describe the utility of assessment tools and imaging methods for the evaluation of CI, and propose a comprehensive diagnostic and management approach.
Collapse
Affiliation(s)
- Sotiria Liori
- Heart Failure Unit and University Clinic of Emergency Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece.
| | - Angelos Arfaras-Melainis
- Heart Failure Unit and University Clinic of Emergency Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Vasiliki Bistola
- Heart Failure Unit and University Clinic of Emergency Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Eftihia Polyzogopoulou
- Heart Failure Unit and University Clinic of Emergency Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - John Parissis
- Heart Failure Unit and University Clinic of Emergency Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| |
Collapse
|
13
|
Li J, Wang K, Huang B, Li R, Wang X, Zhang H, Tang H, Chen X. The receptor for advanced glycation end products mediates dysfunction of airway epithelial barrier in a lipopolysaccharides-induced murine acute lung injury model. Int Immunopharmacol 2021; 93:107419. [PMID: 33548580 DOI: 10.1016/j.intimp.2021.107419] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 01/10/2021] [Accepted: 01/18/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Airway epithelial cells (AECs) act as the first barrier protecting against invasion of environment agents and maintain integrity of lung structure and function. Dysfunction of airway epithelial barrier has been shown to be involved in ALI/ARDS pathogenesis. Yet, the exact mechanism is still obscure. Our study evaluated whether the receptor for advanced glycation end products (RAGE) mediates impaired airway epithelial barrier in LPS-induced murine ALI model. METHODS Male BALB/c mice were subjected to intratracheal instillation of LPS to generate an ALI model. Inhibitors of RAGE, FPS-ZM1 and Azeliragon were respectively given to the mice through intraperitoneal injection. Bronchoalveolar lavage fluid (BALF) and lung tissues were collected for further analysis. RESULTS LPS exposure led to markedly increased expression of RAGE and its ligands HMGB1, HSP70, S100b. Treatment of FPS-ZM1 or Azeliragon not only effectively descended the expression of RAGE and its ligands but also attenuated LPS-induced neutrophil-predominant airway inflammation and injury, decreased levels of IL-6, IL-1β and TNF-α in BALF, alleviated increased alveolar-capillary permeability and pulmonary edema. LPS stimulation significantly impaired the integrity of airway epithelium, paralleled with dislocation of adheren junction (AJ) protein E-cadherin at cell-cell contacts and down-expression of both AJ and tight junction (TJ) proteins Claudin-2 and occludin, all of which were dramatically rescued by RAGE inhibition. CONCLUSION RAGE signaling mediates airway epithelial barrier dysfunction in a LPS-induced ALI murine model.
Collapse
Affiliation(s)
- Jiahui Li
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China
| | - Kai Wang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China
| | - Bo Huang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China
| | - Rui Li
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China
| | - Xilong Wang
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China
| | - Hailing Zhang
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China
| | - Haixiong Tang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China.
| | - Xin Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China.
| |
Collapse
|
14
|
Yang L, Liu Y, Wang Y, Li J, Liu N. Azeliragon ameliorates Alzheimer's disease via the Janus tyrosine kinase and signal transducer and activator of transcription signaling pathway. Clinics (Sao Paulo) 2021; 76:e2348. [PMID: 33681944 PMCID: PMC7920406 DOI: 10.6061/clinics/2021/e2348] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 11/05/2020] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVES TTP488, an antagonist of the receptor for advanced glycation end-products, was evaluated as a potential treatment for patients with mild-to-moderate Alzheimer's disease (AD). However, the mechanism underlying the protective action of TTP488 against AD has not yet been fully explored. METHODS Healthy male rats were exposed to aberrant amyloid β (Aβ) 1-42. Lipopolysaccharide (LPS) and the NOD-like receptor family pyrin domain containing 1 (NLRP1) overexpression lentivirus were injected to activate the NLRP1 inflammasome and exacerbate AD. TTP488 was administered to reverse AD injury. Finally, tofacitinib and fludarabine were used to inhibit the activity of Janus tyrosine kinase (JAK) and signal transducer and activator of transcription (STAT) to prove the relationship between the JAK/STAT signaling pathway and TTP488. RESULTS LPS and NLRP1 overexpression significantly increased the NLRP1 levels, reduced neurological function, and aggravated neuronal damage, as demonstrated by the impact latency time of, time spent by, and length of the platform covered by, the mice in the Morris water maze assay, Nissl staining, and immunofluorescence staining in rats with AD. CONCLUSIONS TTP488 administration successfully reduced AD injury and reversed the aforementioned processes. Additionally, tofacitinib and fludarabine administration could further reverse AD injury after the TTP488 intervention. These results suggest a new potential mechanism underlying the TTP488-mediated alleviation of AD injury.
Collapse
Affiliation(s)
- Lijuan Yang
- Nursing Faculty of Xingtai Medical College, Xingtai, Hebei 054008, China
| | - Yepei Liu
- Medical Image Center, Xingtai City Fifth Hospital, Xingtai, Hebei 054008, China
| | - Yuanyuan Wang
- Nursing Faculty of Xingtai Medical College, Xingtai, Hebei 054008, China
| | - Junsheng Li
- Nursing Faculty of Xingtai Medical College, Xingtai, Hebei 054008, China
- *Corresponding authors. E-mails: /
| | - Na Liu
- Nursing Faculty of Xingtai Medical College, Xingtai, Hebei 054008, China
- *Corresponding authors. E-mails: /
| |
Collapse
|
15
|
Oh Y, Do HTT, Kim S, Kim YM, Chin YW, Cho J. Memory-Enhancing Effects of Mangosteen Pericarp Water Extract through Antioxidative Neuroprotection and Anti-Apoptotic Action. Antioxidants (Basel) 2020; 10:antiox10010034. [PMID: 33396950 PMCID: PMC7823671 DOI: 10.3390/antiox10010034] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/24/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023] Open
Abstract
Mangosteen has long been utilized as a traditional medicine in Southeast Asia. Diverse extracts of mangosteen pericarp and its bioactive xanthones exhibit various bioactivities. However, the pharmacological potential of mangosteen pericarp water extract (MPW) has not been reported yet. This study used primary cultured rat cortical cells to investigate the effect of MPW on neurotoxicity. We found that MPW inhibited neurotoxicity and production of reactive oxygen species triggered by Aβ(25–35) or excitatory amino acids. MPW inhibited caspase 3 activation and DNA fragmentation in Aβ(25–35)- or N-methyl-D-aspartate-treated cells, suggesting an anti-apoptotic action. Additionally, MPW reduced lipid peroxidation and scavenged 1,1-diphenyl-2-picrylhydrazyl radicals, assuring its antioxidant property. Furthermore, MPW suppressed β-secretase and acetylcholinesterase activities. These findings prompted us to evaluate its effect on memory dysfunction in scopolamine-treated mice using Morris water maze test. Oral administration of MPW at the dosage of 50, 100, or 300 mg/kg for four days significantly decreased the latency time to find the platform and markedly increased the swimming time in the target quadrant. Taken together, our results suggest that MPW exerts memory-enhancing effect through antioxidative neuroprotection and anti-apoptotic action. Accordingly, MPW may have a potential to prevent or treat memory impairment associated with Alzheimer’s disease.
Collapse
Affiliation(s)
- Yeonsoo Oh
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Dongguk-ro 32, Ilsandong-gu, Goyang, Gyeonggi 10326, Korea; (Y.O.); (H.T.T.D.); (S.K.)
| | - Ha Thi Thu Do
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Dongguk-ro 32, Ilsandong-gu, Goyang, Gyeonggi 10326, Korea; (Y.O.); (H.T.T.D.); (S.K.)
| | - Sunyoung Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Dongguk-ro 32, Ilsandong-gu, Goyang, Gyeonggi 10326, Korea; (Y.O.); (H.T.T.D.); (S.K.)
| | - Young-Mi Kim
- College of Pharmacy, Seoul National University, Seoul 08826, Korea; (Y.-M.K.); (Y.-W.C.)
| | - Young-Won Chin
- College of Pharmacy, Seoul National University, Seoul 08826, Korea; (Y.-M.K.); (Y.-W.C.)
| | - Jungsook Cho
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Dongguk-ro 32, Ilsandong-gu, Goyang, Gyeonggi 10326, Korea; (Y.O.); (H.T.T.D.); (S.K.)
- Correspondence: ; Tel.: +82-31-961-5211
| |
Collapse
|
16
|
Ettcheto M, Busquets O, Espinosa-Jiménez T, Verdaguer E, Auladell C, Camins A. A Chronological Review of Potential Disease-Modifying Therapeutic Strategies for Alzheimer's Disease. Curr Pharm Des 2020; 26:1286-1299. [PMID: 32066356 DOI: 10.2174/1381612826666200211121416] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 12/18/2019] [Indexed: 01/28/2023]
Abstract
Late-onset Alzheimer's disease (LOAD) is a neurodegenerative disorder that has become a worldwide health problem. This pathology has been classically characterized for its affectation on cognitive function and the presence of depositions of extracellular amyloid β-protein (Aβ) and intracellular neurofibrillary tangles (NFT) composed of hyperphosphorylated Tau protein. To this day, no effective treatment has been developed. Multiple strategies have been proposed over the years with the aim of finding new therapeutic approaches, such as the sequestration of Aβ in plasma or the administration of anti-inflammatory drugs. Also, given the significant role of the insulin receptor in the brain in the proper maintenance of cognitive function, drugs focused on the amelioration of insulin resistance have been proposed as potentially useful and effective in the treatment of AD. In the present review, taking into account the molecular complexity of the disease, it has been proposed that the most appropriate therapeutic strategy is a combinatory treatment of several drugs that will regulate a wide spectrum of the described altered pathological pathways.
Collapse
Affiliation(s)
- Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Department of Biochemistry and Biotechnology, Faculty of Medicine and Life Sciences, University Rovira i Virgili, Reus, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Oriol Busquets
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Department of Biochemistry and Biotechnology, Faculty of Medicine and Life Sciences, University Rovira i Virgili, Reus, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Triana Espinosa-Jiménez
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Ester Verdaguer
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Carme Auladell
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, Madrid, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| |
Collapse
|
17
|
Hall D. A simple method for modeling amyloid kinetics featuring position biased fiber breakage. Biophys Physicobiol 2020; 17:30-35. [PMID: 33110736 PMCID: PMC7550252 DOI: 10.2142/biophysico.bsj-2020003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 03/12/2020] [Indexed: 12/01/2022] Open
Abstract
A mathematical model of amyloid fiber formation is described that is able to simply specify different rates of fiber breakage at internal versus end regions. This Note presents the derivation of the relevant equations and provides results showing the dramatic effects of position biased fiber breakage on the kinetics of amyloid growth.
Collapse
Affiliation(s)
- Damien Hall
- Laboratory of Biochemistry and Genetics, NIDDK, NIH, Bethesda, MD 20892-0830, USA.,Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan.,Present address: International Center, Nagoya Institute of Technology, Gokiso, Showa, Nagoya, Aichi 466-8555, Japan
| |
Collapse
|
18
|
Martins M, Silva R, M. M. Pinto M, Sousa E. Marine Natural Products, Multitarget Therapy and Repurposed Agents in Alzheimer's Disease. Pharmaceuticals (Basel) 2020; 13:E242. [PMID: 32933034 PMCID: PMC7558913 DOI: 10.3390/ph13090242] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/01/2020] [Accepted: 09/07/2020] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is a multifactorial disease characterized by the presence of amyloid plaques, neurofibrillary tangles, and nerve cell death that affects, mainly, older people. After decades of investigation, the search for an efficacious treatment for AD remains and several strategies can be and are being employed in this journey. In this review, four of the most promising strategies, alongside with its most promising agents under investigation or development are highlighted. Marine natural products (MNP) are a source of unique chemical structures with useful biological activities for AD treatment. One of the most promising compounds, a marine-derived acidic oligosaccharide (GV-971) just passed phase III clinical trials with a unique mechanism of action. Combination therapy and multitargeted-directed ligand therapy (MTDL) are also two important strategies, with several examples in clinical trials, based on the belief that the best approach for AD is a therapy capable of modulating multiple target pathways. Drug repurposing, a strategy that requires a smaller investment and is less time consuming, is emerging as a strong contender with a variety of pharmacological agents resurfacing in an attempt to identify a therapeutic candidate capable of modifying the course of this disease.
Collapse
Affiliation(s)
- Márcia Martins
- CIIMAR—Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal; (M.M.); (M.M.M.P.)
- Laboratório de Química Orgânica e Farmacêutica, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Renata Silva
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal;
| | - Madalena M. M. Pinto
- CIIMAR—Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal; (M.M.); (M.M.M.P.)
- Laboratório de Química Orgânica e Farmacêutica, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Emília Sousa
- CIIMAR—Centro Interdisciplinar de Investigação Marinha e Ambiental, Terminal de Cruzeiros do Porto de Leixões, 4450-208 Matosinhos, Portugal; (M.M.); (M.M.M.P.)
- Laboratório de Química Orgânica e Farmacêutica, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| |
Collapse
|
19
|
Lozupone M, Solfrizzi V, D'Urso F, Di Gioia I, Sardone R, Dibello V, Stallone R, Liguori A, Ciritella C, Daniele A, Bellomo A, Seripa D, Panza F. Anti-amyloid-β protein agents for the treatment of Alzheimer's disease: an update on emerging drugs. Expert Opin Emerg Drugs 2020; 25:319-335. [PMID: 32772738 DOI: 10.1080/14728214.2020.1808621] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Currently available Alzheimer's disease (AD) therapeutics are only symptomatic, targeting cholinergic and glutamatergic neurotransmissions. Several putative disease-modifying drugs in late-stage clinical development target amyloid-β (Aβ) peptide and tau protein, the principal neurophatological hallmarks of the disease. AREAS COVERED Phase III randomized clinical trials of anti-Aβ drugs for AD treatment were searched in US and EU clinical trial registries and principal biomedical databases until May 2020. EXPERT OPINION At present, compounds in Phase III clinical development for AD include four anti-Ab monoclonal antibodies (solanezumab, gantenerumab, aducanumab, BAN2401), the combination of cromolyn sodium and ibuprofen (ALZT-OP1), and two small molecules (levetiracetam, GV-971). These drugs are mainly being tested in subjects during early AD phases or at preclinical stage of familial AD or even in asymptomatic subjects at high risk of developing AD. The actual results support the hypothesis that elevated Aβ represents an early stage in the AD continuum and demonstrate the feasibility of enrolling these high-risk participants in secondary prevention trials to slow cognitive decline during the AD preclinical stages. However, a series of clinical failures may question further development of Aβ-targeting drugs and the findings from current ongoing Phase III trials will hopefully give light to this critical issue.
Collapse
Affiliation(s)
- Madia Lozupone
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro , Bari, Italy
| | - Vincenzo Solfrizzi
- "Cesare Frugoni" Internal and Geriatric Medicine and Memory Unit, University of Bari "Aldo Moro" , Bari, Italy
| | - Francesca D'Urso
- Psychiatric Unit, Department of Clinical and Experimental Medicine, University of Foggia , Foggia, Italy
| | - Ilaria Di Gioia
- Psychiatric Unit, Department of Clinical and Experimental Medicine, University of Foggia , Foggia, Italy
| | - Rodolfo Sardone
- Population Health Unit - "Salus in Apulia Study" - National Institute of Gastroenterology, "Saverio De Bellis", Research Hospital , Bari, Italy
| | - Vittorio Dibello
- Population Health Unit - "Salus in Apulia Study" - National Institute of Gastroenterology, "Saverio De Bellis", Research Hospital , Bari, Italy.,Department of Orofacial Pain and Dysfunction, Academic Centre of Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam , The Netherlands
| | - Roberta Stallone
- Population Health Unit - "Salus in Apulia Study" - National Institute of Gastroenterology, "Saverio De Bellis", Research Hospital , Bari, Italy
| | - Angelo Liguori
- Population Health Unit - "Salus in Apulia Study" - National Institute of Gastroenterology, "Saverio De Bellis", Research Hospital , Bari, Italy
| | - Chiara Ciritella
- Physical and Rehabilitation Medicine Department, University of Foggia , Foggia, Italy
| | - Antonio Daniele
- Institute of Neurology, Catholic University of Sacred Heart , Rome, Italy.,Institute of Neurology, Fondazione Policlinico Universitario A. Gemelli IRCCS , Rome, Italy
| | - Antonello Bellomo
- Psychiatric Unit, Department of Clinical and Experimental Medicine, University of Foggia , Foggia, Italy
| | - Davide Seripa
- Geriatric Unit and Gerontology-Geriatrics Research Laboratory, Department of Medical Sciences, IRCCS Casa Sollievo Della Sofferenza , Foggia, Italy.,Hematology and Stem Cell Transplant Unit, Vito Fazzi Hospital, ASL Lecce , Lecce, Italy
| | - Francesco Panza
- Population Health Unit - "Salus in Apulia Study" - National Institute of Gastroenterology, "Saverio De Bellis", Research Hospital , Bari, Italy
| |
Collapse
|
20
|
De Martinis M, Ginaldi L, Sirufo MM, Pioggia G, Calapai G, Gangemi S, Mannucci C. Alarmins in Osteoporosis, RAGE, IL-1, and IL-33 Pathways: A Literature Review. MEDICINA (KAUNAS, LITHUANIA) 2020; 56:medicina56030138. [PMID: 32204562 PMCID: PMC7142770 DOI: 10.3390/medicina56030138] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/10/2020] [Accepted: 03/12/2020] [Indexed: 12/13/2022]
Abstract
Alarmins are endogenous mediators released by cells following insults or cell death to alert the host’s innate immune system of a situation of danger or harm. Many of these, such as high-mobility group box-1 and 2 (HMGB1, HMGB2) and S100 (calgranulin proteins), act through RAGE (receptor for advanced glycation end products), whereas the IL-1 and IL-33 cytokines bind the IL-1 receptors type I and II, and the cellular receptor ST2, respectively. The alarmin family and their signal pathways share many similarities of cellular and tissue localization, functions, and involvement in various physiological processes and inflammatory diseases including osteoporosis. The aim of the review was to evaluate the role of alarmins in osteoporosis. A bibliographic search of the published scientific literature regarding the role of alarmins in osteoporosis was organized independently by two researchers in the following scientific databases: Pubmed, Scopus, and Web of Science. The keywords used were combined as follows: “alarmins and osteoporosis”, “RAGE and osteoporosis”, “HMGB1 and osteoporosis”, “IL-1 and osteoporosis”, “IL 33 and osteopororsis”, “S100s protein and osteoporosis”. The information was summarized and organized in the present review. We highlight the emerging roles of alarmins in various bone remodeling processes involved in the onset and development of osteoporosis, as well as their potential role as biomarkers of osteoporosis severity and progression. Findings of the research suggest a potential use of alarmins as pharmacological targets in future therapeutic strategies aimed at preventing bone loss and fragility fractures induced by aging and inflammatory diseases.
Collapse
Affiliation(s)
- Massimo De Martinis
- Department of Life, Health, & Environmental Sciences, University of L’Aquila, 6700 L’Aquila, Italy; (M.D.M.); (L.G.); (M.M.S.)
| | - Lia Ginaldi
- Department of Life, Health, & Environmental Sciences, University of L’Aquila, 6700 L’Aquila, Italy; (M.D.M.); (L.G.); (M.M.S.)
| | - Maria Maddalena Sirufo
- Department of Life, Health, & Environmental Sciences, University of L’Aquila, 6700 L’Aquila, Italy; (M.D.M.); (L.G.); (M.M.S.)
| | - Giovanni Pioggia
- National Research Council of Italy (CNR)-Institute for Biomedical Research and Innovation (IRIB), 98164 Messina, Italy;
| | - Gioacchino Calapai
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy;
| | - Sebastiano Gangemi
- School and Division of Allergy and Clinical Immunology, Department of Experimental Medicine, University of Messina, 98125 Messina, Italy;
| | - Carmen Mannucci
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy;
- Correspondence: ; Tel.: +39-090-22-12-697
| |
Collapse
|
21
|
Kwan P, Konno H, Chan KY, Baum L. Rationale for the development of an Alzheimer's disease vaccine. Hum Vaccin Immunother 2020; 16:645-653. [PMID: 31526227 PMCID: PMC7227628 DOI: 10.1080/21645515.2019.1665453] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 08/13/2019] [Accepted: 09/02/2019] [Indexed: 02/07/2023] Open
Abstract
Vaccination traditionally has targeted infectious agents and thus has not heretofore been used to prevent neurodegenerative illness. However, amyloid β (Aβ) or tau, which can act like infectious proteins, or prions, might induce Alzheimer's disease (AD). Furthermore, evidence suggests that traditional infectious agents, including certain viruses and bacteria, may trigger AD. It is therefore worth exploring whether removing such targets could prevent AD. Although failing to treat AD patients who already display cognitive impairment, Aβ monoclonal antibodies are being tested in pre-symptomatic, at-risk individuals to prevent dementia. These antibodies might become the first AD therapeutics. However, their high cost will keep them out of the arms of the vast majority of patients, who increasingly live in developing countries. Because vaccines produce antibodies internally at much lower cost, vaccination might be the most promising approach to reducing the global burden of dementia.
Collapse
Affiliation(s)
- Ping Kwan
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, P.R. China
- Sydney School of Veterinary Science, The University of Sydney, Sydney, Australia
| | - Haruki Konno
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, P.R. China
| | - Ka Yan Chan
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, P.R. China
| | - Larry Baum
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, P.R. China
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, P.R. China
- Center for Genomic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, P.R. China
- Department of Psychiatry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, P.R. China
| |
Collapse
|
22
|
Panza F, Lozupone M, Dibello V, Greco A, Daniele A, Seripa D, Logroscino G, Imbimbo BP. Are antibodies directed against amyloid-β (Aβ) oligomers the last call for the Aβ hypothesis of Alzheimer's disease? Immunotherapy 2019; 11:3-6. [PMID: 30702009 DOI: 10.2217/imt-2018-0119] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Affiliation(s)
- Francesco Panza
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, & Sense Organs, University of Bari Aldo Moro, Bari, Italy.,Department of ClinicalResearch in Neurology, Center for Neurodegenerative Diseases and the AgingBrain, University of Bari "Aldo Moro", "Pia Fondazione Cardinale G. Panico", Tricase, Lecce, Italy.,Geriatric Unit, Fondazione IRCCS "Casa Sollievo della Sofferenza", SanGiovanni Rotondo, Foggia, Italy
| | - Madia Lozupone
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, & Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Vittorio Dibello
- Interdisciplinary Department of Medicine (DIM), Section of Dentistry, University of Bari Aldo Moro, Bari, Italy
| | - Antonio Greco
- Geriatric Unit, Fondazione IRCCS "Casa Sollievo della Sofferenza", SanGiovanni Rotondo, Foggia, Italy
| | - Antonio Daniele
- Institute of Neurology, Catholic University of Sacred Heart, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Davide Seripa
- Geriatric Unit, Fondazione IRCCS "Casa Sollievo della Sofferenza", SanGiovanni Rotondo, Foggia, Italy
| | - Giancarlo Logroscino
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, & Sense Organs, University of Bari Aldo Moro, Bari, Italy.,Department of ClinicalResearch in Neurology, Center for Neurodegenerative Diseases and the AgingBrain, University of Bari "Aldo Moro", "Pia Fondazione Cardinale G. Panico", Tricase, Lecce, Italy
| | - Bruno P Imbimbo
- Department of Researchand Development, Chiesi Farmaceutici, Parma, Italy
| |
Collapse
|
23
|
Paudel YN, Angelopoulou E, Jones NC, O’Brien TJ, Kwan P, Piperi C, Othman I, Shaikh MF. Tau Related Pathways as a Connecting Link between Epilepsy and Alzheimer's Disease. ACS Chem Neurosci 2019; 10:4199-4212. [PMID: 31532186 DOI: 10.1021/acschemneuro.9b00460] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Emerging findings point toward an important interconnection between epilepsy and Alzheimer's disease (AD) pathogenesis. Patients with epilepsy (PWE) commonly exhibit cognitive impairment similar to AD patients, who in turn are at a higher risk of developing epilepsy compared to age-matched controls. To date, no disease-modifying treatment strategy is available for either epilepsy or AD, reflecting an immediate need for exploring common molecular targets, which can delineate a possible mechanistic link between epilepsy and AD. This review attempts to disentangle the interconnectivity between epilepsy and AD pathogenesis via the crucial contribution of Tau protein. Tau protein is a microtubule-associated protein (MAP) that has been implicated in the pathophysiology of both epilepsy and AD. Hyperphosphorylation of Tau contributes to the different forms of human epilepsy and inhibition of the same exerted seizure inhibitions and altered disease progression in a range of animal models. Moreover, Tau-protein-mediated therapy has demonstrated promising outcomes in experimental models of AD. In this review, we discuss how Tau-related mechanisms might present a link between the cause of seizures in epilepsy and cognitive disruption in AD. Untangling this interconnection might be instrumental in designing novel therapies that can minimize epileptic seizures and cognitive deficits in patients with epilepsy and AD.
Collapse
Affiliation(s)
- Yam Nath Paudel
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 46150, Malaysia
| | - Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens 10679, Greece
| | - Nigel C. Jones
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne 3800, Australia
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Royal Parade, Parkville, Victoria 3010, Australia
| | - Terence J. O’Brien
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne 3800, Australia
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Royal Parade, Parkville, Victoria 3010, Australia
| | - Patrick Kwan
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne 3800, Australia
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Royal Parade, Parkville, Victoria 3010, Australia
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens 10679, Greece
| | - Iekhsan Othman
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 46150, Malaysia
| | - Mohd. Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 46150, Malaysia
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne 3800, Australia
| |
Collapse
|
24
|
Muensterman ET, Luo Y, Parker JM. Breakthrough Therapy, PRIME and Sakigake: A Comparison Between Neuroscience and Oncology in Obtaining Preferred Regulatory Status. Ther Innov Regul Sci 2019:2168479019874062. [PMID: 31558045 DOI: 10.1177/2168479019874062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Because of the increasing demand for drugs addressing life-threatening and rare diseases, regulatory agencies have developed a variety of accelerated regulatory pathways. These programs are aimed at prioritizing the most promising drug candidates for diseases lacking satisfactory treatments. The most prominent accelerated programs introduced have been Breakthrough-Therapy Designation (BTD) in the United States, Priority Medicine (PRIME) in the European Union and Sakigake in Japan. This article reviews these designations and looks at differences in how they are granted across the 3 jurisdictions focusing on neuroscience and oncology. METHODS Our objective was to analyze BTD, PRIME, and Sakigake approvals between 2012 and 2019 with a focus on numerical disparities of designations granted between the 2 therapeutic areas. A search of public sources pertaining to topics of BTD, PRIME, and Sakigake was undertaken. RESULTS This analysis revealed that 48% of BTD were granted in oncology, while neuroscience received 8% of these designations, for PRIME designations were 27% received by oncology and 15% by neuroscience and in Japan, 50% of Sakigake were granted to oncology and 22% to neuroscience products. CONCLUSION Given the global nature of drug development and relative similarity of these regulatory mechanisms, there is an apparent disparity between the US granting special status at 6:1 (oncology: neuroscience) and both the EU and Japan granting at 2:1. This disproportionate ratio is likely impacted by multifactorial issues; however, this difference is worth further investigation.
Collapse
Affiliation(s)
| | - Yijia Luo
- Takeda Pharmaceutical Company, Cambridge, MA, USA
| | | |
Collapse
|
25
|
Staffaroni AM, Ljubenkov PA, Kornak J, Cobigo Y, Datta S, Marx G, Walters SM, Chiang K, Olney N, Elahi FM, Knopman DS, Dickerson BC, Boeve BF, Gorno-Tempini ML, Spina S, Grinberg LT, Seeley WW, Miller BL, Kramer JH, Boxer AL, Rosen HJ. Longitudinal multimodal imaging and clinical endpoints for frontotemporal dementia clinical trials. Brain 2019; 142:443-459. [PMID: 30698757 DOI: 10.1093/brain/awy319] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/02/2018] [Indexed: 12/22/2022] Open
Abstract
Frontotemporal dementia refers to a group of progressive neurodegenerative syndromes usually caused by the accumulation of pathological tau or TDP-43 proteins. The effects of these proteins in the brain are complex, and each can present with several different clinical syndromes. Clinical efficacy trials of drugs targeting these proteins must use endpoints that are meaningful to all participants despite the variability in symptoms across patients. There are many candidate clinical measures, including neuropsychological scores and functional measures. Brain imaging is another potentially attractive outcome that can be precisely quantified and provides evidence of disease modification. Most imaging studies in frontotemporal dementia have been cross-sectional, and few have compared longitudinal changes in cortical volume with changes in other measures such as perfusion and white matter integrity. The current study characterized longitudinal changes in 161 patients with three frontotemporal dementia syndromes: behavioural variant frontotemporal dementia (n = 77) and the semantic (n = 45) and non-fluent (n = 39) variants of primary progressive aphasia. Visits included comprehensive neuropsychological and functional assessment, structural MRI (3 T), diffusion tensor imaging, and arterial spin labelled perfusion imaging. The goal was to identify measures that are appropriate as clinical trial outcomes for each group, as well as those that might be appropriate for trials that would include more than one of these groups. Linear mixed effects models were used to estimate changes in each measure, and to examine the correlation between imaging and clinical changes. Sample sizes were estimated based on the observed effects for theoretical clinical trials using bootstrapping techniques to provide 95% confidence intervals for these estimates. Declines in functional and neuropsychological measures, as well as frontal and temporal cortical volumes and white matter microstructure were detected in all groups. Imaging changes were statistically significantly correlated with, and explained a substantial portion of variance in, the change in most clinical measures. Perfusion and diffusion tensor imaging accounted for variation in clinical decline beyond volume alone. Sample size estimates for atrophy and diffusion imaging were comparable to clinical measures. Corpus callosal fractional anisotropy led to the lowest sample size estimates for all three syndromes. These findings provide further guidance on selection of trial endpoints for studies in frontotemporal dementia and support the use of neuroimaging, particularly structural and diffusion weighted imaging, as biomarkers. Diffusion and perfusion imaging appear to offer additional utility for explaining clinical change beyond the variance explained by volume alone, arguing for considering multimodal imaging in treatment trials.
Collapse
Affiliation(s)
- Adam M Staffaroni
- Department of Neurology, Memory and Aging Center, University of California at San Francisco (UCSF), San Francisco, CA, USA
| | - Peter A Ljubenkov
- Department of Neurology, Memory and Aging Center, University of California at San Francisco (UCSF), San Francisco, CA, USA
| | - John Kornak
- Department of Epidemiology and Biostatistics, University of California at San Francisco (UCSF), San Francisco, CA, USA
| | - Yann Cobigo
- Department of Neurology, Memory and Aging Center, University of California at San Francisco (UCSF), San Francisco, CA, USA
| | - Samir Datta
- Department of Neurology, Memory and Aging Center, University of California at San Francisco (UCSF), San Francisco, CA, USA
| | - Gabe Marx
- Department of Neurology, Memory and Aging Center, University of California at San Francisco (UCSF), San Francisco, CA, USA
| | - Samantha M Walters
- Department of Neurology, Memory and Aging Center, University of California at San Francisco (UCSF), San Francisco, CA, USA
| | - Kevin Chiang
- Department of Neurology, Memory and Aging Center, University of California at San Francisco (UCSF), San Francisco, CA, USA
| | - Nick Olney
- Department of Neurology, Memory and Aging Center, University of California at San Francisco (UCSF), San Francisco, CA, USA
| | - Fanny M Elahi
- Department of Neurology, Memory and Aging Center, University of California at San Francisco (UCSF), San Francisco, CA, USA
| | - David S Knopman
- Department of Neurology, College of Medicine, Mayo Clinic, 200 First Street SW, Rochester, Minnesota USA
| | - Bradford C Dickerson
- Frontotemporal Disorders Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charleston, MA, USA
| | - Bradley F Boeve
- Department of Neurology, College of Medicine, Mayo Clinic, 200 First Street SW, Rochester, Minnesota USA
| | - Maria Luisa Gorno-Tempini
- Department of Neurology, Memory and Aging Center, University of California at San Francisco (UCSF), San Francisco, CA, USA
| | - Salvatore Spina
- Department of Neurology, Memory and Aging Center, University of California at San Francisco (UCSF), San Francisco, CA, USA
| | - Lea T Grinberg
- Department of Neurology, Memory and Aging Center, University of California at San Francisco (UCSF), San Francisco, CA, USA.,Department of Pathology, Memory and Aging Center, University of California at San Francisco (UCSF), San Francisco, CA, USA.,Department of Pathology - LIM 22, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - William W Seeley
- Department of Neurology, Memory and Aging Center, University of California at San Francisco (UCSF), San Francisco, CA, USA.,Department of Pathology, Memory and Aging Center, University of California at San Francisco (UCSF), San Francisco, CA, USA
| | - Bruce L Miller
- Department of Neurology, Memory and Aging Center, University of California at San Francisco (UCSF), San Francisco, CA, USA
| | - Joel H Kramer
- Department of Neurology, Memory and Aging Center, University of California at San Francisco (UCSF), San Francisco, CA, USA
| | - Adam L Boxer
- Department of Neurology, Memory and Aging Center, University of California at San Francisco (UCSF), San Francisco, CA, USA
| | - Howard J Rosen
- Department of Neurology, Memory and Aging Center, University of California at San Francisco (UCSF), San Francisco, CA, USA
| |
Collapse
|
26
|
Yu D, Yan H, Zhou J, Yang X, Lu Y, Han Y. A circuit view of deep brain stimulation in Alzheimer's disease and the possible mechanisms. Mol Neurodegener 2019; 14:33. [PMID: 31395077 PMCID: PMC6688355 DOI: 10.1186/s13024-019-0334-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/26/2019] [Indexed: 02/08/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by chronic progressive cognitive deterioration frequently accompanied by psychopathological symptoms, including changes in personality and social isolation, which severely reduce quality of life. Currently, no viable therapies or present-day drugs developed for the treatment of AD symptoms are able to slow or reverse AD progression or prevent the advance of neurodegeneration. As such, non-drug alternatives are currently being tested, including deep brain stimulation (DBS). DBS is an established therapy for several neurological and psychiatric indications, such as movement disorders. Studies assessing DBS for other disorders have also found improvements in cognitive function, providing the impetus for clinical trials on DBS for AD. Targets of DBS in AD clinical trials and animal model studies include the fornix, entorhinal cortex (EC), nucleus basalis of Meynert (NBM), and vertical limb of diagonal band (VDB). However, there is still no comprehensive theory explaining the effects of DBS on AD symptoms or a consensus on which targets provide optimal benefits. This article reviews the anatomy of memory circuits related to AD, as well as studies on DBS rescue of AD in these circuits and the possible therapeutic mechanisms.
Collapse
Affiliation(s)
- Danfang Yu
- Department of Neurobiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Neurology, Provincial Hospital of Integrated Chinese & Western Medicine, Wuhan, China
| | - Huanhuan Yan
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Biomedical Engineering Department, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Zhou
- Department of Neurobiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaodan Yang
- Department of Neurobiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Youming Lu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. .,Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China.
| | - Yunyun Han
- Department of Neurobiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. .,Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
27
|
Davis HM, Essex AL, Valdez S, Deosthale PJ, Aref MW, Allen MR, Bonetto A, Plotkin LI. Short-term pharmacologic RAGE inhibition differentially affects bone and skeletal muscle in middle-aged mice. Bone 2019; 124:89-102. [PMID: 31028960 PMCID: PMC6543548 DOI: 10.1016/j.bone.2019.04.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/17/2019] [Accepted: 04/22/2019] [Indexed: 12/27/2022]
Abstract
Loss of bone and muscle mass are two major clinical complications among the growing list of chronic diseases that primarily affect elderly individuals. Persistent low-grade inflammation, one of the major drivers of aging, is also associated with both bone and muscle dysfunction in aging. Particularly, chronic activation of the receptor for advanced glycation end products (RAGE) and elevated levels of its ligands high mobility group box 1 (HMGB1), AGEs, S100 proteins and Aβ fibrils have been linked to bone and muscle loss in various pathologies. Further, genetic or pharmacologic RAGE inhibition has been shown to preserve both bone and muscle mass. However, whether short-term pharmacologic RAGE inhibition can prevent early bone and muscle loss in aging is unknown. To address this question, we treated young (4-mo) and middle-aged (15-mo) C57BL/6 female mice with vehicle or Azeliragon, a small-molecule RAGE inhibitor initially developed to treat Alzheimer's disease. Azeliragon did not prevent the aging-induced alterations in bone geometry or mechanics, likely due to its differential effects [direct vs. indirect] on bone cell viability/function. On the other hand, Azeliragon attenuated the aging-related body composition changes [fat and lean mass] and reversed the skeletal muscle alterations induced with aging. Interestingly, while Azeliragon induced similar metabolic changes in bone and skeletal muscle, aging differentially altered the expression of genes associated with glucose uptake/metabolism in these two tissues, highlighting a potential explanation for the differential effects of Azeliragon on bone and skeletal muscle in middle-aged mice. Overall, our findings suggest that while short-term pharmacologic RAGE inhibition did not protect against early aging-induced bone alterations, it prevented against the early effects of aging in skeletal muscle.
Collapse
Affiliation(s)
- Hannah M Davis
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States of America; Indiana Center for Musculoskeletal Health, Indianapolis, IN, United States of America.
| | - Alyson L Essex
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States of America; Indiana Center for Musculoskeletal Health, Indianapolis, IN, United States of America.
| | - Sinai Valdez
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States of America.
| | - Padmini J Deosthale
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States of America; Indiana Center for Musculoskeletal Health, Indianapolis, IN, United States of America.
| | - Mohammad W Aref
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States of America; Indiana Center for Musculoskeletal Health, Indianapolis, IN, United States of America.
| | - Matthew R Allen
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States of America; Indiana Center for Musculoskeletal Health, Indianapolis, IN, United States of America; Roudebush Veterans Administration Medical Center, Indianapolis, IN, United States of America.
| | - Andrea Bonetto
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States of America; Indiana Center for Musculoskeletal Health, Indianapolis, IN, United States of America; Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States of America.
| | - Lilian I Plotkin
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States of America; Indiana Center for Musculoskeletal Health, Indianapolis, IN, United States of America; Roudebush Veterans Administration Medical Center, Indianapolis, IN, United States of America.
| |
Collapse
|
28
|
Angehrn Z, Nordon C, Turner A, Gove D, Karcher H, Keenan A, Neumann M, Sostar J, de Reydet de Vulpillieres F. Ethical and social implications of using predictive modeling for Alzheimer's disease prevention: a systematic literature review protocol. BMJ Open 2019; 9:e026468. [PMID: 30833325 PMCID: PMC6443073 DOI: 10.1136/bmjopen-2018-026468] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 01/03/2019] [Accepted: 01/08/2019] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION The therapeutic paradigm in Alzheimer's disease (AD) has shifted towards secondary prevention, defined as an intervention aiming to prevent or delay disease onset in pre-symptomatic individuals at risk of developing dementia due to AD. The key feature of AD prevention is the need to treat years or even decades before the onset of cognitive, behavioural or functional decline. Prediction of AD risk and evaluation of long-term treatment outcomes in this setting requires predictive modelling and is associated with ethical concerns and social implications. The objective of this review is to identify and elucidate them, as presented in the literature. METHODS AND ANALYSIS A systematic literature review was conducted in Medline, Embase, PsycInfo and Scopus, and was complemented with a grey literature search. All searches were conducted between March and July 2018. Two reviewers independently assessed each study for inclusion and disagreements were adjudicated by a third reviewer. Data are now being extracted using an extraction sheet developed within the group of reviewers, based on an initial sample of three manuscripts, but allowing for inclusion of newly identified data items (ethical arguments). Data will be analysed qualitatively using a thematic analysis technique. Potential biases in selection and interpretation of extracted data are mitigated by the fact that reviewers come from a range of different scientific backgrounds and represent different types of stakeholders in this ethical discussion (academia, industry, patient advocacy groups). ETHICS AND DISSEMINATION The study does not require ethical approval. The findings of the review will be disseminated in a peer-reviewed journal and presented at conferences. They will also be reported through the Innovative Medicine Initiative project: Real World Outcomes Across the AD Spectrum for Better Care: Multi-modal Data Access Platform (IMI: ROADMAP). TRIAL REGISTRATION NUMBER CRD42018092205.
Collapse
Affiliation(s)
| | | | - Andrew Turner
- Oxford Internet Institute, University of Oxford, Oxford, UK
| | | | | | | | | | - Jelena Sostar
- Analytica Laser, a Certara Company, Loerrach, Germany
| | | |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW The receptor for advanced glycation end products (RAGE) and several of its ligands have been implicated in the onset and progression of pathologies associated with aging, chronic inflammation, and cellular stress. In particular, the role of RAGE and its ligands in bone tissue during both physiological and pathological conditions has been investigated. However, the extent to which RAGE signaling regulates bone homeostasis and disease onset remains unclear. Further, RAGE effects in the different bone cells and whether these effects are cell-type specific is unknown. The objective of the current review is to describe the literature over RAGE signaling in skeletal biology as well as discuss the clinical potential of RAGE as a diagnostic and/or therapeutic target in bone disease. RECENT FINDINGS The role of RAGE and its ligands during skeletal homeostasis, tissue repair, and disease onset/progression is beginning to be uncovered. For example, detrimental effects of the RAGE ligands, advanced glycation end products (AGEs), have been identified for osteoblast viability/activity, while others have observed that low level AGE exposure stimulates osteoblast autophagy, which subsequently promotes viability and function. Similar findings have been reported with HMGB1, another RAGE ligand, in which high levels of the ligand are associated with osteoblast/osteocyte apoptosis, whereas low level/short-term administration stimulates osteoblast differentiation/bone formation and promotes fracture healing. Additionally, elevated levels of several RAGE ligands (AGEs, HMGB1, S100 proteins) induce osteoblast/osteocyte apoptosis and stimulate cytokine production, which is associated with increased osteoclast differentiation/activity. Conversely, direct RAGE-ligand exposure in osteoclasts may have inhibitory effects. These observations support a conclusion that elevated bone resorption observed in conditions of high circulating ligands and RAGE expression are due to actions on osteoblasts/osteocytes rather than direct actions on osteoclasts, although additional work is required to substantiate the observations. Recent studies have demonstrated that RAGE and its ligands play an important physiological role in the regulation of skeletal development, homeostasis, and repair/regeneration. Conversely, elevated levels of RAGE and its ligands are clearly related with various diseases associated with increased bone loss and fragility. However, despite the recent advancements in the field, many questions regarding RAGE and its ligands in skeletal biology remain unanswered.
Collapse
Affiliation(s)
- Lilian I Plotkin
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Dr., MS 5023, Indianapolis, IN, 46202, USA.
- Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA.
- Indiana Center for Musculoskeletal Health, Indianapolis, IN, USA.
| | - Alyson L Essex
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Dr., MS 5023, Indianapolis, IN, 46202, USA
- Indiana Center for Musculoskeletal Health, Indianapolis, IN, USA
| | - Hannah M Davis
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Dr., MS 5023, Indianapolis, IN, 46202, USA
- Indiana Center for Musculoskeletal Health, Indianapolis, IN, USA
| |
Collapse
|
30
|
Gagliardi S, Franco V, Sorrentino S, Zucca S, Pandini C, Rota P, Bernuzzi S, Costa A, Sinforiani E, Pansarasa O, Cashman JR, Cereda C. Curcumin and Novel Synthetic Analogs in Cell-Based Studies of Alzheimer's Disease. Front Pharmacol 2018; 9:1404. [PMID: 30559668 PMCID: PMC6287206 DOI: 10.3389/fphar.2018.01404] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/15/2018] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder that is associated with the most common type of dementia and is characterized by the presence of deposits of the protein fragment amyloid beta (Aβ) in the brain. The natural product mixture of curcuminoids that improves certain defects in innate immune cells of AD patients may selectively enhance Aβ phagocytosis by alteration of gene transcription. In this work, we evaluated the protective effects of curcuminoids in cells from AD patients by investigating the effect on NF-κB and BACE1 signaling pathways. These results were compared to the gene expression profile of the clearance of Aβ. The minor curcumin constituent, bisdemethoxycurcumin (BDC) showed the most potent protective action to decrease levels of NF-κB and BACE1, decrease the inflammatory cascade and diminish Aβ aggregates in cells from AD patients. Moreover, mannosyl-glycoprotein 4-beta-N-acetylglucosaminyltransferase (MGAT3) and vitamin D receptor (VDR) gene mRNAs were up-regulated in peripheral blood mononuclear cells from AD patients treated with BDC. BDC treatment impacts both gene expression including Mannosyl (Beta-1,4-)-Glycoprotein Beta-1,4-N-Acetylglucosaminyltransferase, Vitamin D and Toll like receptor mRNA and Aβ phagocytosis. The observation of down-regulation of BACE1 and NF-κB following administration of BDC to cells from AD patients as a model system may have utility in the treatment of asymptomatic AD patients.
Collapse
Affiliation(s)
- Stella Gagliardi
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Valentina Franco
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy
- Clinical and Experimental Pharmacology Unit, Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | | | - Susanna Zucca
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Cecilia Pandini
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Pavia, Italy
| | - Paola Rota
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Stefano Bernuzzi
- Immunohematological and Transfusional Service, Centre of Transplantation Immunology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Alfredo Costa
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Neurology Department, IRCCS Mondino Foundation, Pavia, Italy
| | - Elena Sinforiani
- Laboratory of Neuropsychology/UVA, IRCCS Mondino Foundation, Pavia, Italy
| | - Orietta Pansarasa
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy
| | - John R. Cashman
- Laboratory of Chemistry, Human BioMolecular Research Institute, San Diego, CA, United States
| | - Cristina Cereda
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
31
|
Solfrizzi V, Agosti P, Lozupone M, Custodero C, Schilardi A, Valiani V, Santamato A, Sardone R, Dibello V, Di Lena L, Stallone R, Ranieri M, Bellomo A, Greco A, Daniele A, Seripa D, Sabbà C, Logroscino G, Panza F. Nutritional interventions and cognitive-related outcomes in patients with late-life cognitive disorders: A systematic review. Neurosci Biobehav Rev 2018; 95:480-498. [PMID: 30395922 DOI: 10.1016/j.neubiorev.2018.10.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/27/2018] [Accepted: 10/29/2018] [Indexed: 12/30/2022]
Abstract
There have been a large number of observational studies on the impact of nutrition on neuroprotection, however, there was a lack of evidence from randomized clinical trials (RCTs). In the present systematic review, from the 32 included RCTs published in the last four years (2014-2017) in patients aged 60 years and older with different late-life cognitive disorders, nutritional intervention through medical food/nutraceutical supplementation and multidomain approach improved magnetic resonance imaging findings and other cognitive-related biomarkers, but without clear effect on cognition in mild Alzheimer's disease (AD) and mild cognitive impairment (MCI). Antioxidant-rich foods (nuts, grapes, cherries) and fatty acid supplementation, mainly n-3 polyunsaturated fatty acids (PUFA), improved specific cognitive domains and cognitive-related outcomes in MCI, mild-to-moderate dementia, and AD. Antioxidant vitamin and trace element supplementations improved only cognitive-related outcomes and biomarkers, high-dose B vitamin supplementation in AD and MCI patients improved cognitive outcomes in the subjects with a high baseline plasma n-3 PUFA, while folic acid supplementation had positive impact on specific cognitive domains in those with high homocysteine.
Collapse
Affiliation(s)
- Vincenzo Solfrizzi
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari 'Aldo Moro', Bari, Italy.
| | - Pasquale Agosti
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari 'Aldo Moro', Bari, Italy
| | - Madia Lozupone
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Carlo Custodero
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari 'Aldo Moro', Bari, Italy
| | - Andrea Schilardi
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari 'Aldo Moro', Bari, Italy
| | - Vincenzo Valiani
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari 'Aldo Moro', Bari, Italy
| | - Andrea Santamato
- Physical Medicine and Rehabilitation Section, "OORR Hospital", University of Foggia, Foggia, Italy
| | - Rodolfo Sardone
- National Institute of Gastroenterology "Saverio de Bellis", Research Hospital, Castellana Grotte, Bari, Italy; Interdisciplinary Department of Medicine (DIM), Section of Dentistry, University of Bari AldoMoro, Bari, Italy
| | - Vittorio Dibello
- Psychiatric Unit, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Luca Di Lena
- National Institute of Gastroenterology "Saverio de Bellis", Research Hospital, Castellana Grotte, Bari, Italy; Interdisciplinary Department of Medicine (DIM), Section of Dentistry, University of Bari AldoMoro, Bari, Italy
| | - Roberta Stallone
- National Institute of Gastroenterology "Saverio de Bellis", Research Hospital, Castellana Grotte, Bari, Italy; Interdisciplinary Department of Medicine (DIM), Section of Dentistry, University of Bari AldoMoro, Bari, Italy
| | - Maurizio Ranieri
- Physical Medicine and Rehabilitation Section, "OORR Hospital", University of Foggia, Foggia, Italy
| | - Antonello Bellomo
- Psychiatric Unit, Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Antonio Greco
- Geriatric Unit, Fondazione IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy
| | - Antonio Daniele
- Institute of Neurology, Catholic University of Sacred Heart, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Davide Seripa
- Geriatric Unit, Fondazione IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy
| | - Carlo Sabbà
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari 'Aldo Moro', Bari, Italy
| | - Giancarlo Logroscino
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy; Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, University of Bari "Aldo Moro", "Pia Fondazione Cardinale G. Panico", Tricase, Lecce, Italy
| | - Francesco Panza
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy; Geriatric Unit, Fondazione IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Foggia, Italy; Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, University of Bari "Aldo Moro", "Pia Fondazione Cardinale G. Panico", Tricase, Lecce, Italy.
| |
Collapse
|
32
|
Lim EW, Aarsland D, Ffytche D, Taddei RN, van Wamelen DJ, Wan YM, Tan EK, Ray Chaudhuri K. Amyloid-β and Parkinson's disease. J Neurol 2018; 266:2605-2619. [PMID: 30377818 DOI: 10.1007/s00415-018-9100-8] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/16/2022]
Abstract
Parkinson's disease (PD) is the second commonest neurodegenerative disorder in the world with a rising prevalence. The pathophysiology is multifactorial but aggregation of misfolded α-synuclein is considered to be a key underpinning mechanism. Amyloid-β (Aβ) and tau deposition are also comorbid associations and especially Aβ deposition is associated with cognitive decline in PD. Some existing evidence suggests that low cerebrospinal fluid (CSF) Aβ42 is predictive of future cognitive impairment in PD. Recent studies also show that CSF Aβ is associated with the postural instability and gait difficulties (PIGD) or the newly proposed cholinergic subtype of PD, a possible risk factor for cognitive decline in PD. The glial-lymphatic system, responsible for convective solute clearance driven by active fluid transport through aquaporin-4 water channels, may be implicated in brain amyloid deposition. A better understanding of the role of this system and more specifically the role of Aβ in PD symptomatology, could introduce new treatment and repurposing drug-based strategies. For instance, apomorphine infusion has been shown to promote the degradation of Aβ in rodent models. This is further supported in a post-mortem study in PD patients although clinical implications are unclear. In this review, we address the clinical implication of cerebral Aβ deposition in PD and elaborate on its metabolism, its role in cognition and motor function/gait, and finally assess the potential effect of apomorphine on Aβ deposition in PD.
Collapse
Affiliation(s)
- Ee Wei Lim
- Parkinson Foundation International Centre of Excellence at King's College Hospital, Denmark Hill, London, SE5 9RS, UK. .,Department of Neurology, National Neuroscience Institute (Singapore General Hospital Campus), 20 College Road, Singapore, 169856, Singapore. .,Duke-National University of Singapore Graduate Medical School, Singapore, 169857, Singapore.
| | - Dag Aarsland
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience at King's College London, De Crespigny Park, London, SE5 8AF, UK
| | - Dominic Ffytche
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience at King's College London, De Crespigny Park, London, SE5 8AF, UK
| | - Raquel Natalia Taddei
- Parkinson Foundation International Centre of Excellence at King's College Hospital, Denmark Hill, London, SE5 9RS, UK
| | - Daniel J van Wamelen
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience at King's College London, De Crespigny Park, London, SE5 8AF, UK.,Parkinson Foundation International Centre of Excellence at King's College Hospital, Denmark Hill, London, SE5 9RS, UK.,Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Centre, Reinier Postlaan 4, Postbus 9101, 6500HB, Nijmegen, The Netherlands
| | - Yi-Min Wan
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience at King's College London, De Crespigny Park, London, SE5 8AF, UK.,Parkinson Foundation International Centre of Excellence at King's College Hospital, Denmark Hill, London, SE5 9RS, UK.,Department of Psychiatry, Ng Teng Fong General Hospital, 1 Jurong East Street 21, Singapore, 609606, Singapore
| | - Eng King Tan
- Department of Neurology, National Neuroscience Institute (Singapore General Hospital Campus), 20 College Road, Singapore, 169856, Singapore.,Duke-National University of Singapore Graduate Medical School, Singapore, 169857, Singapore
| | - Kallol Ray Chaudhuri
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience at King's College London, De Crespigny Park, London, SE5 8AF, UK.,Parkinson Foundation International Centre of Excellence at King's College Hospital, Denmark Hill, London, SE5 9RS, UK
| | | |
Collapse
|
33
|
New hope for Alzheimer's dementia as prospects for disease modification fade: symptomatic treatments for agitation and psychosis. CNS Spectr 2018; 23:291-297. [PMID: 30382934 DOI: 10.1017/s1092852918001360] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
More than a decade of failure to find disease-modifying treatments for Alzheimer's dementia has driven the field back to finding better symptomatic treatments for behavioral symptoms of dementia, especially psychosis and agitation.
Collapse
|
34
|
Zhang Y, Huai Y, Zhang X, Song C, Cai J, Zhang Y. The Mode of Action of an Anti-Oligomeric Amyloid β-Protein Antibody Affects its Protective Efficacy. Neurotox Res 2018; 35:304-317. [PMID: 30229545 DOI: 10.1007/s12640-018-9955-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 09/06/2018] [Accepted: 09/07/2018] [Indexed: 12/22/2022]
Abstract
The process of developing antibody drugs for Alzheimer's disease therapy has been both long and difficult; however, recent advances suggest that antibodies against neurotoxic Αβ42 can suppress the progression of AD, especially on its early stage. Here, we obtained and characterized a novel anti-oligomeric Aβ42 aggregate scFv antibody, HT7, which could induce the significant disaggregation of Aβ42 aggregates through the release of stable and non-cytotoxic hexameric complexes that were composed of three scFv HT7s and one Aβ42 trimer, the latter being found to serve as the assembled subunit within larger Aβ42 aggregates in addition to existing freely between the cells. The docking model of the scFv HT7-Aβ42 complex revealed that only the N-terminal peptide of the Aβ42 molecule was bound into the groove between the VH and VL domains of scFv HT7. Thus, it was suggested that the hydrophobic interaction between the C-terminal peptides of Aβ42 molecules maintained the stability of the Aβ42 trimers or the Aβ42 trimer subunits. The saturation of Aβ42 trimer subunits by scFv HT7 and the subsequent dissociation of the scFv HT7-saturated Aβ42 trimer subunits from larger Aβ42 aggregates constituted the primary mechanisms underlying the high efficacy of scFv HT7. Our findings revealed that it was not sufficient for an anti-oligomeric Aβ42 antibody to exhibit high specificity and high affinity to the oligomeric Aβ42 aggregates in order to promote Aβ42 aggregate clearance and neutralize their cytotoxic effects. Here, for the first time, we proposed a "post-saturation dissociation" mechanism of Aβ42 oligomeric subunits for effective anti-Aβ42 antibodies.
Collapse
Affiliation(s)
- Yunlong Zhang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, 2699 Qian-Jin Street, Changchun, 130012, Jilin, China
| | - Yangyang Huai
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, 2699 Qian-Jin Street, Changchun, 130012, Jilin, China
| | - Xiaoning Zhang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, 2699 Qian-Jin Street, Changchun, 130012, Jilin, China
| | - Chuli Song
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, 2699 Qian-Jin Street, Changchun, 130012, Jilin, China
| | - Jing Cai
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, 2699 Qian-Jin Street, Changchun, 130012, Jilin, China.,China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yingjiu Zhang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, 2699 Qian-Jin Street, Changchun, 130012, Jilin, China. .,School of Life Science, Jilin University, Changchun, China.
| |
Collapse
|
35
|
VanPatten S, Al-Abed Y. High Mobility Group Box-1 (HMGb1): Current Wisdom and Advancement as a Potential Drug Target. J Med Chem 2018; 61:5093-5107. [PMID: 29268019 DOI: 10.1021/acs.jmedchem.7b01136] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
High mobility group box-1 (HMGb1) protein, a nuclear non-histone protein that is released or secreted from the cell in response to damage or stress, is a sentinel for the immune system that plays a critical role in cell survival/death pathways. This review highlights key features of the endogenous danger-associated molecular pattern (DAMP) protein, HMGb1 in the innate inflammatory response along with various cofactors and receptors that regulate its downstream effects. The evidence demonstrating increased levels of HMGb1 in human inflammatory diseases and conditions is presented, along with a summary of current small molecule or peptide-like antagonists proven to specifically target HMGb1. Additionally, we delineate the measures needed toward validating this protein as a clinically relevant biomarker or bioindicator and as a relevant drug target.
Collapse
Affiliation(s)
- Sonya VanPatten
- Center for Molecular Innovation , The Feinstein Institute for Medical Research , 350 Community Drive , Manhasset , New York 11030 , United States
| | - Yousef Al-Abed
- Center for Molecular Innovation , The Feinstein Institute for Medical Research , 350 Community Drive , Manhasset , New York 11030 , United States
| |
Collapse
|
36
|
Lee HJ, Savelieff MG, Kang J, Brophy MB, Nakashige TG, Lee SJC, Nolan EM, Lim MH. Calprotectin influences the aggregation of metal-free and metal-bound amyloid-β by direct interaction. Metallomics 2018; 10:1116-1127. [DOI: 10.1039/c8mt00091c] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
CP-Ser [S100A8(C42S)/S100A9(C3S) oligomer] interacts with metal-free and metal-bound Aβ40 peptides and modulates their aggregation in the absence and presence of metal ions.
Collapse
Affiliation(s)
- Hyuck Jin Lee
- Department of Chemistry
- Korea Advanced Institute of Science and Technology (KAIST)
- Daejeon 34141
- Republic of Korea
| | | | - Juhye Kang
- Department of Chemistry
- Korea Advanced Institute of Science and Technology (KAIST)
- Daejeon 34141
- Republic of Korea
- Department of Chemistry
| | | | | | - Shin Jung C. Lee
- Department of Chemistry
- Ulsan National Institute of Science and Technology (UNIST)
- Ulsan 44919
- Republic of Korea
| | - Elizabeth M. Nolan
- Department of Chemistry
- Massachusetts Institute of Technology (MIT)
- Cambridge
- USA
| | - Mi Hee Lim
- Department of Chemistry
- Korea Advanced Institute of Science and Technology (KAIST)
- Daejeon 34141
- Republic of Korea
| |
Collapse
|
37
|
Panza F, Seripa D, Lozupone M, Solfrizzi V, Imbimbo BP, Barulli MR, Tortelli R, Capozzo R, Bisceglia P, Dimitri A, Stallone R, Dibello V, Quaranta N, Daniele A, Bellomo A, Greco A, Logroscino G. The potential of solanezumab and gantenerumab to prevent Alzheimer’s disease in people with inherited mutations that cause its early onset. Expert Opin Biol Ther 2017; 18:25-35. [DOI: 10.1080/14712598.2018.1389885] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Francesco Panza
- Unit of Neurodegenerative Disease, Department of Basic Medicine Sciences, Neuroscience, and Sense Organs, University of Bari ‘Aldo Moro’, Bari, Italy
- Unit of Neurodegenerative Disease, Department of Clinical Research in Neurology, University of Bari ‘Aldo Moro’ at ‘Pia Fondazione Card. G. Panico’, Tricase, Italy
- Geriatric Unit and Gerontology-Geriatrics Research Laboratory, Department of Medical Sciences, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Davide Seripa
- Geriatric Unit and Gerontology-Geriatrics Research Laboratory, Department of Medical Sciences, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Madia Lozupone
- Unit of Neurodegenerative Disease, Department of Basic Medicine Sciences, Neuroscience, and Sense Organs, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Vincenzo Solfrizzi
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Bruno P. Imbimbo
- Research & Development Department, Chiesi Farmaceutici, Parma, Italy
| | - Maria Rosaria Barulli
- Unit of Neurodegenerative Disease, Department of Clinical Research in Neurology, University of Bari ‘Aldo Moro’ at ‘Pia Fondazione Card. G. Panico’, Tricase, Italy
| | - Rosanna Tortelli
- Unit of Neurodegenerative Disease, Department of Clinical Research in Neurology, University of Bari ‘Aldo Moro’ at ‘Pia Fondazione Card. G. Panico’, Tricase, Italy
| | - Rosa Capozzo
- Unit of Neurodegenerative Disease, Department of Clinical Research in Neurology, University of Bari ‘Aldo Moro’ at ‘Pia Fondazione Card. G. Panico’, Tricase, Italy
| | - Paola Bisceglia
- Geriatric Unit and Gerontology-Geriatrics Research Laboratory, Department of Medical Sciences, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Andrea Dimitri
- Psychiatric Unit, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Roberta Stallone
- Unit of Neurodegenerative Disease, Department of Basic Medicine Sciences, Neuroscience, and Sense Organs, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Vittorio Dibello
- Interdisciplinary Department of Medicine (DIM), Section of Dentistry, University of Bari Aldo, Moro, Italy
| | - Nicola Quaranta
- Otolaryngology Unit, University of Bari Aldo Moro, Bari, Italy
| | - Antonio Daniele
- Institute of Neurology, Catholic University of Sacred Heart, Rome, Italy
| | - Antonello Bellomo
- Psychiatric Unit, Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari ‘Aldo Moro’, Bari, Italy
| | - Antonio Greco
- Geriatric Unit and Gerontology-Geriatrics Research Laboratory, Department of Medical Sciences, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Giancarlo Logroscino
- Unit of Neurodegenerative Disease, Department of Basic Medicine Sciences, Neuroscience, and Sense Organs, University of Bari ‘Aldo Moro’, Bari, Italy
- Unit of Neurodegenerative Disease, Department of Clinical Research in Neurology, University of Bari ‘Aldo Moro’ at ‘Pia Fondazione Card. G. Panico’, Tricase, Italy
| |
Collapse
|
38
|
van Raalte DH, Verchere CB. Improving glycaemic control in type 2 diabetes: Stimulate insulin secretion or provide beta-cell rest? Diabetes Obes Metab 2017; 19:1205-1213. [PMID: 28295962 DOI: 10.1111/dom.12935] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 03/06/2017] [Accepted: 03/08/2017] [Indexed: 12/25/2022]
Abstract
Type 2 diabetes (T2D) is characterized by a gradual decline in pancreatic beta cell function that determines the progressive course of the disease. While beta-cell failure is an important contributor to hyperglycaemia, chronic hyperglycaemia itself is also detrimental for beta-cell function, probably by inducing prolonged secretory stress on the beta cell as well as through direct glucotoxic mechanisms that have not been fully defined. For years, research has been carried out in search of therapies targeting hyperglycaemia that preserve long-term beta-cell function in T2D, a quest that is still ongoing. Current strategies aim to improve glycaemic control, either by promoting endogenous insulin secretion, such as sulfonylureas, or by mechanisms that may impact the beta cell indirectly, for example, providing beta-cell rest through insulin treatment. Although overall long-term success is limited with currently available interventions, in this review we argue that strategies that induce beta-cell rest have considerable potential to preserve long-term beta-cell function. This is based on laboratory-based studies involving human islets as well as clinical studies employing intensive insulin therapy, thiazolidinediones, bariatric surgery, short-acting glucagon-like peptide (GLP)-1 receptor agonists and a promising new class of diabetes drugs, sodium-glucose-linked transporter (SGLT)-2 inhibitors. Nevertheless, a lack of long-term clinical studies that focus on beta-cell function for the newer glucose-lowering agents, as well as commonly used combination therapies, preclude a straightforward conclusion; this gap in our knowledge should be a focus of future studies.
Collapse
Affiliation(s)
- Daniël H van Raalte
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, Amsterdam, The Netherlands
- Department of Surgery, The University of British Columbia and Research Institute, BC Children's Hospital, Vancouver, Canada
| | - C Bruce Verchere
- Department of Surgery, The University of British Columbia and Research Institute, BC Children's Hospital, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia and Research Institute, BC Children's Hospital, Vancouver, Canada
| |
Collapse
|
39
|
Chierrito TPC, Pedersoli-Mantoani S, Roca C, Requena C, Sebastian-Perez V, Castillo WO, Moreira NCS, Pérez C, Sakamoto-Hojo ET, Takahashi CS, Jiménez-Barbero J, Cañada FJ, Campillo NE, Martinez A, Carvalho I. From dual binding site acetylcholinesterase inhibitors to allosteric modulators: A new avenue for disease-modifying drugs in Alzheimer's disease. Eur J Med Chem 2017; 139:773-791. [PMID: 28863358 DOI: 10.1016/j.ejmech.2017.08.051] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/21/2017] [Accepted: 08/22/2017] [Indexed: 12/31/2022]
Abstract
The lack of an effective treatment for Alzheimer' disease (AD), an increasing prevalence and severe neurodegenerative pathology boost medicinal chemists to look for new drugs. Currently, only acethylcholinesterase (AChE) inhibitors and glutamate antagonist have been approved to the palliative treatment of AD. Although they have a short-term symptomatic benefits, their clinical use have revealed important non-cholinergic functions for AChE such its chaperone role in beta-amyloid toxicity. We propose here the design, synthesis and evaluation of non-toxic dual binding site AChEIs by hybridization of indanone and quinoline heterocyclic scaffolds. Unexpectely, we have found a potent allosteric modulator of AChE able to target cholinergic and non-cholinergic functions by fixing a specific AChE conformation, confirmed by STD-NMR and molecular modeling studies. Furthermore the promising biological data obtained on human neuroblastoma SH-SY5Y cell assays for the new allosteric hybrid 14, led us to propose it as a valuable pharmacological tool for the study of non-cholinergic functions of AChE, and as a new important lead for novel disease modifying agents against AD.
Collapse
Affiliation(s)
- Talita P C Chierrito
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café, s/n, 14040-903, Ribeirão Preto, SP, Brazil
| | - Susimaire Pedersoli-Mantoani
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café, s/n, 14040-903, Ribeirão Preto, SP, Brazil
| | - Carlos Roca
- IPSBB Unit, Centro de Investigaciones Biologicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Carlos Requena
- IPSBB Unit, Centro de Investigaciones Biologicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Victor Sebastian-Perez
- IPSBB Unit, Centro de Investigaciones Biologicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Willian O Castillo
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirão Preto, SP, Brazil
| | - Natalia C S Moreira
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirão Preto, SP, Brazil
| | - Concepción Pérez
- Instituto de Química Médica (CSIC), Juan de la Cierva 3, 28006, Madrid, Spain
| | - Elza T Sakamoto-Hojo
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirão Preto, SP, Brazil; Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Avenida Bandeirantes, 3900, 14040-900, Ribeirão Preto, SP, Brazil
| | - Catarina S Takahashi
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av. Bandeirantes, 3900, 14049-900, Ribeirão Preto, SP, Brazil; Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Avenida Bandeirantes, 3900, 14040-900, Ribeirão Preto, SP, Brazil
| | - Jesús Jiménez-Barbero
- IPSBB Unit, Centro de Investigaciones Biologicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain; CIC BioGUNE, Parque Tecnologico de Bizkaia, Edif. 801A, 48160, Derio-Bizkaia, Bilbao, Spain
| | - F Javier Cañada
- IPSBB Unit, Centro de Investigaciones Biologicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Nuria E Campillo
- IPSBB Unit, Centro de Investigaciones Biologicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Ana Martinez
- IPSBB Unit, Centro de Investigaciones Biologicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain.
| | - Ivone Carvalho
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Avenida do Café, s/n, 14040-903, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
40
|
Solfrizzi V, Custodero C, Lozupone M, Imbimbo BP, Valiani V, Agosti P, Schilardi A, D’Introno A, La Montagna M, Calvani M, Guerra V, Sardone R, Abbrescia DI, Bellomo A, Greco A, Daniele A, Seripa D, Logroscino G, Sabbá C, Panza F. Relationships of Dietary Patterns, Foods, and Micro- and Macronutrients with Alzheimer’s Disease and Late-Life Cognitive Disorders: A Systematic Review. J Alzheimers Dis 2017; 59:815-849. [DOI: 10.3233/jad-170248] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Vincenzo Solfrizzi
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari Aldo Moro, Bari, Italy
| | - Carlo Custodero
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari Aldo Moro, Bari, Italy
| | - Madia Lozupone
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Bruno P. Imbimbo
- Department of Research and Development, Chiesi Farmaceutici, Parma, Italy
| | - Vincenzo Valiani
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari Aldo Moro, Bari, Italy
| | - Pasquale Agosti
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari Aldo Moro, Bari, Italy
| | - Andrea Schilardi
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari Aldo Moro, Bari, Italy
| | - Alessia D’Introno
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari Aldo Moro, Bari, Italy
| | - Maddalena La Montagna
- Department of Clinical and Experimental Medicine, Psychiatric Unit, University of Foggia, Foggia, Italy
| | - Mariapaola Calvani
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Vito Guerra
- National Institute for Digestive Diseases, IRCCS “Saverio de Bellis”, Castellana, Bari, Italy
| | - Rodolfo Sardone
- National Institute for Digestive Diseases, IRCCS “Saverio de Bellis”, Castellana, Bari, Italy
| | - Daniela I. Abbrescia
- National Institute for Digestive Diseases, IRCCS “Saverio de Bellis”, Castellana, Bari, Italy
| | - Antonello Bellomo
- Department of Clinical and Experimental Medicine, Psychiatric Unit, University of Foggia, Foggia, Italy
| | - Antonio Greco
- Geriatric Unit and Laboratory of Gerontology and Geriatrics, Department of Medical Sciences, IRCCS “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, Foggia, Italy
| | - Antonio Daniele
- Institute of Neurology, Catholic University of Sacred Heart, Rome, Italy
| | - Davide Seripa
- Geriatric Unit and Laboratory of Gerontology and Geriatrics, Department of Medical Sciences, IRCCS “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, Foggia, Italy
| | - Giancarlo Logroscino
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
- Department of Clinical Research in Neurology, University of Bari Aldo Moro, “Pia Fondazione Cardinale G. Panico”, Tricase, Lecce, Italy
| | - Carlo Sabbá
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Panza
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
- Geriatric Unit and Laboratory of Gerontology and Geriatrics, Department of Medical Sciences, IRCCS “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, Foggia, Italy
- Department of Clinical Research in Neurology, University of Bari Aldo Moro, “Pia Fondazione Cardinale G. Panico”, Tricase, Lecce, Italy
| |
Collapse
|
41
|
Does treating hearing loss prevent or slow the progress of dementia? Hearing is not all in the ears, but who's listening? CNS Spectr 2017; 22:247-250. [PMID: 28376938 DOI: 10.1017/s1092852917000268] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Since hearing loss is strongly linked to cognitive decline, treating hearing impairment may have the potential to prevent the onset or slow the progress of dementia.
Collapse
|
42
|
Translation of imaging biomarkers from clinical research to healthcare. Z Gerontol Geriatr 2017; 50:84-88. [DOI: 10.1007/s00391-017-1225-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 03/10/2017] [Accepted: 03/13/2017] [Indexed: 01/12/2023]
|
43
|
Puchowicz MA, Seyfried TN. Novel ketone body therapy for managing Alzheimer's disease: An Editorial Highlight for Effects of a dietary ketone ester on hippocampal glycolytic and tricarboxylic acid cycle intermediates and amino acids in a 3xTgAD mouse model of Alzheimer's disease. J Neurochem 2017; 141:162-164. [PMID: 28299805 DOI: 10.1111/jnc.13979] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 01/26/2017] [Accepted: 01/30/2017] [Indexed: 11/29/2022]
Abstract
Read the highlighted article 'Effects of a dietary ketone ester on hippocampal glycolytic and tricarboxylic acid cycle intermediates and amino acids in a 3xTgAD mouse model of Alzheimer's disease' on page 195.
Collapse
Affiliation(s)
- Michelle A Puchowicz
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Thomas N Seyfried
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA
| |
Collapse
|
44
|
Hunter S, Brayne C. Do anti-amyloid beta protein antibody cross reactivities confound Alzheimer disease research? J Negat Results Biomed 2017; 16:1. [PMID: 28126004 PMCID: PMC5270220 DOI: 10.1186/s12952-017-0066-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/13/2017] [Indexed: 12/20/2022] Open
Abstract
Background Alzheimer disease (AD) research has focussed mainly on the amyloid beta protein (Aβ). However, many Aβ-and P3-type peptides derived from the amyloid precursor protein (APP) and peptides thought to derive from Aβ catabolism share sequence homology. Additionally, conformations can change dependent on aggregation state and solubility leading to significant uncertainty relating to interpretations of immunoreactivity with antibodies raised against Aβ. We review evidence relating to the reactivities of commonly used antibodies including 6F3D, 6E10 and 4G8 and evaluate their reactivity profiles with respect to AD diagnosis and research. Results Antibody cross-reactivities between Aβ-type, P3-type and Aβ-catabolic peptides confound interpretations of immunoreactivity. More than one antibody is required to adequately characterise Aβ. The relationships between anti-Aβ immunoreactivity, neuropathology and proposed APP cleavages are unclear. Conclusions We find that the concept of Aβ lacks clarity as a specific entity. Anti-Aβ antibody cross-reactivities lead to significant uncertainty in our understanding of the APP proteolytic system and its role in AD with profound implications for current research and therapeutic strategies.
Collapse
Affiliation(s)
- Sally Hunter
- Department of Public Health and Primary Care, Institute of Public Health Forvie Site, University of Cambridge School of Clinical Medicine, Box 113 Cambridge Biomedical Campus, Cambridge, CB2 0SP, UK.
| | - Carol Brayne
- Department of Public Health and Primary Care, Institute of Public Health Forvie Site, University of Cambridge School of Clinical Medicine, Box 113 Cambridge Biomedical Campus, Cambridge, CB2 0SP, UK
| |
Collapse
|
45
|
Song Y, Kim HD, Lee MK, Hong IH, Won CK, Bai HW, Lee SS, Lee S, Chung BY, Cho JH. Maysin and Its Flavonoid Derivative from Centipedegrass Attenuates Amyloid Plaques by Inducting Humoral Immune Response with Th2 Skewed Cytokine Response in the Tg (APPswe, PS1dE9) Alzheimer's Mouse Model. PLoS One 2017; 12:e0169509. [PMID: 28072821 PMCID: PMC5224976 DOI: 10.1371/journal.pone.0169509] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 12/18/2016] [Indexed: 01/01/2023] Open
Abstract
Alzheimer’s disease (AD) is a slow, progressive neurodegenerative disease and the most common type of dementia in the elderly. The etiology of AD and its underlying mechanism are still not clear. In a previous study, we found that an ethyl acetate extract of Centipedegrass (CG) (i.e., EA-CG) contained 4 types of Maysin derivatives, including Luteolin, Isoorientin, Rhamnosylisoorientin, and Derhamnosylmaysin, and showed protective effects against Amyloid beta (Aβ) by inhibiting oligomeric Aβ in cellular and in vitro models. Here, we examined the preventative effects of EA-CG treatment on the Aβ burden in the Tg (Mo/Hu APPswe PS1dE9) AD mouse model. We have investigated the EA-CG efficacy as novel anti-AD likely preventing amyloid plaques using immunofluorescence staining to visually analyze Aβ40/42 and fibril formation with Thioflavin-S or 6E10 which are the profile of immunoreactivity against epitope Aβ1–16 or neuritic plaque, the quantitation of humoral immune response against Aβ, and the inflammatory cytokine responses (Th1 and Th2) using ELISA and QRT-PCR. To minimize the toxicity of the extracted CG, we addressed the liver toxicity in response to the CG extract treatment in Tg mice using relevant markers, such as aspartate aminotransferase (AST)/ alanine aminotransferase (ALT) measurements in serum. The EA-CG extract significantly reduced the Aβ burden, the concentration of soluble Aβ40/42 protein, and fibril formation in the hippocampus and cortex of the Tg mice treated with EA-CG (50 mg/kg BW/day) for 6 months compared with the Tg mice treated with a normal diet. Additionally, the profile of anti-inflammatory cytokines revealed that the levels of Th2 (interleukin-4 (IL-4) and interleukin-10 (IL-10)) cytokines are more significantly increased than Th1 (interferon-γ (IFN-γ), interleukin-2(IL-2)) in the sera. These results suggest that the EA-CG fraction induces IL-4/IL-10-dependent anti-inflammatory cytokines (Th2) rather than pro-inflammatory cytokines (Th1), which are driven by IL-2/IFN-γ. With regard to the immune response, EA-CG induced an immunoglobulin IgG and IgM response against the EA-CG treatment in the Tg mice. Furthermore, EA-CG significantly ameliorated the level of soluble Aβ42 and Aβ40. Similarly, we observed that the fibril formation was also decreased by EA-CG treatment in the hippocampus and cortex after quantitative analysis with Thioflavin-S staining in the Tg brain tissues. Taken together, our findings suggested that Maysin and its derivative flavonoid compounds in the EA-CG fraction might be beneficial therapeutic treatments or alternative preventative measures to adjuvant for boosting humoral and cellular include immune response and anti-inflammation which may lead to amyloid plaque accumulation in Alzheimer’s patients’ brains.
Collapse
Affiliation(s)
- Yuno Song
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, Korea
| | - Hong-Duck Kim
- Department of Environmental Health Science, New York Medical College, Valhalla, New York, United States of America
| | - Min-Kwon Lee
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, Korea
| | - Il-Hwa Hong
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, Korea
| | - Chung-Kil Won
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, Korea
| | - Hyoung-Woo Bai
- Advanced Radiation Technology Institute, Korea Atomic Energy Institute, Jeongeup, Korea
| | - Seung Sik Lee
- Advanced Radiation Technology Institute, Korea Atomic Energy Institute, Jeongeup, Korea
| | - SungBeom Lee
- Advanced Radiation Technology Institute, Korea Atomic Energy Institute, Jeongeup, Korea
| | - Byung Yeoup Chung
- Advanced Radiation Technology Institute, Korea Atomic Energy Institute, Jeongeup, Korea
- * E-mail: (JHC); (BYC)
| | - Jae-Hyeon Cho
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, Korea
- * E-mail: (JHC); (BYC)
| |
Collapse
|
46
|
Panza F, Logroscino G. Anti-tau vaccine in Alzheimer's disease: a tentative step. Lancet Neurol 2016; 16:99-100. [PMID: 27955996 DOI: 10.1016/s1474-4422(16)30340-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 11/09/2016] [Indexed: 01/07/2023]
Affiliation(s)
- Francesco Panza
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari 70100, Italy; Department of Clinical Research in Neurology, University of Bari Aldo Moro, Pia Fondazione Cardinale G Panico, Tricase, Lecce, Italy; Geriatric Unit & Laboratory of Gerontology and Geriatrics, Department of Medical Sciences, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy.
| | - Giancarlo Logroscino
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari 70100, Italy; Department of Clinical Research in Neurology, University of Bari Aldo Moro, Pia Fondazione Cardinale G Panico, Tricase, Lecce, Italy
| |
Collapse
|