1
|
Onisiforou A, Charalambous EG, Zanos P. Shattering the Amyloid Illusion: The Microbial Enigma of Alzheimer's Disease Pathogenesis-From Gut Microbiota and Viruses to Brain Biofilms. Microorganisms 2025; 13:90. [PMID: 39858858 PMCID: PMC11767882 DOI: 10.3390/microorganisms13010090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 12/18/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
For decades, Alzheimer's Disease (AD) research has focused on the amyloid cascade hypothesis, which identifies amyloid-beta (Aβ) as the primary driver of the disease. However, the consistent failure of Aβ-targeted therapies to demonstrate efficacy, coupled with significant safety concerns, underscores the need to rethink our approach to AD treatment. Emerging evidence points to microbial infections as environmental factors in AD pathoetiology. Although a definitive causal link remains unestablished, the collective evidence is compelling. This review explores unconventional perspectives and emerging paradigms regarding microbial involvement in AD pathogenesis, emphasizing the gut-brain axis, brain biofilms, the oral microbiome, and viral infections. Transgenic mouse models show that gut microbiota dysregulation precedes brain Aβ accumulation, emphasizing gut-brain signaling pathways. Viral infections like Herpes Simplex Virus Type 1 (HSV-1) and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) may lead to AD by modulating host processes like the immune system. Aβ peptide's antimicrobial function as a response to microbial infection might inadvertently promote AD. We discuss potential microbiome-based therapies as promising strategies for managing and potentially preventing AD progression. Fecal microbiota transplantation (FMT) restores gut microbial balance, reduces Aβ accumulation, and improves cognition in preclinical models. Probiotics and prebiotics reduce neuroinflammation and Aβ plaques, while antiviral therapies targeting HSV-1 and vaccines like the shingles vaccine show potential to mitigate AD pathology. Developing effective treatments requires standardized methods to identify and measure microbial infections in AD patients, enabling personalized therapies that address individual microbial contributions to AD pathogenesis. Further research is needed to clarify the interactions between microbes and Aβ, explore bacterial and viral interplay, and understand their broader effects on host processes to translate these insights into clinical interventions.
Collapse
Affiliation(s)
- Anna Onisiforou
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus;
- Center of Applied Neuroscience, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus
| | - Eleftheria G. Charalambous
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus;
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, 1–2, Ellernholzstr., 17489 Greifswald, Germany
| | - Panos Zanos
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus;
- Center of Applied Neuroscience, 75 Kallipoleos Avenue, 1678 Nicosia, Cyprus
| |
Collapse
|
2
|
Liu N, Haziyihan A, Zhao W, Chen Y, Chao H. Trajectory of brain-derived amyloid beta in Alzheimer's disease: where is it coming from and where is it going? Transl Neurodegener 2024; 13:42. [PMID: 39160618 PMCID: PMC11331646 DOI: 10.1186/s40035-024-00434-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurological disorder that primarily impacts cognitive function. Currently there are no disease-modifying treatments to stop or slow its progression. Recent studies have found that several peripheral and systemic abnormalities are associated with AD, and our understanding of how these alterations contribute to AD is becoming more apparent. In this review, we focuse on amyloid‑beta (Aβ), a major hallmark of AD, summarizing recent findings on the source of brain-derived Aβ and discussing where and how the brain-derived Aβ is cleared in vivo. Based on these findings, we propose future strategies for AD prevention and treatment, from a novel perspective on Aβ metabolism.
Collapse
Affiliation(s)
- Ni Liu
- Zhengzhou University, Zhengzhou, 450001, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, China
| | | | - Wei Zhao
- Zhengzhou University, Zhengzhou, 450001, China
| | - Yu Chen
- Zhengzhou University, Zhengzhou, 450001, China
| | - Hongbo Chao
- Zhengzhou University, Zhengzhou, 450001, China.
- Huazhong University of Science and Technology, Wuhan, 430074, China.
| |
Collapse
|
3
|
Sharma M, Pal P, Gupta SK. Advances in Alzheimer's disease: A multifaceted review of potential therapies and diagnostic techniques for early detection. Neurochem Int 2024; 177:105761. [PMID: 38723902 DOI: 10.1016/j.neuint.2024.105761] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 04/20/2024] [Accepted: 05/06/2024] [Indexed: 06/04/2024]
Abstract
Alzheimer's disease (AD) remains one of the most formidable neurological disorders, affecting millions globally. This review provides a holistic overview of the therapeutic strategies, both conventional and novel, aimed at mitigating the impact of AD. Initially, we delve into the conventional approach, emphasizing the role of Acetylcholinesterase (AChE) inhibition, which has been a cornerstone in AD management. As our understanding of AD evolves, several novel potential approaches emerge. We discuss the promising roles of Butyrylcholinesterase (BChE) inhibition, Tau Protein inhibitors, COX-2 inhibition, PPAR-γ agonism, and FAHH inhibition, among others. The potential of the endocannabinoids (eCB) system, cholesterol-lowering drugs, metal chelators, and MMPs inhibitors are also explored, culminating in the exploration of the pivotal role of microRNA in AD progression. Parallel to these therapeutic insights, we shed light on the novel tools and methodologies revolutionizing AD research. From the quantitative analysis of gene expression by qRTPCR to the evaluation of mitochondrial function using induced pluripotent stem cells (iPSCs), the advances in diagnostic and research tools offer renewed hope. Moreover, we explore the current landscape of clinical trials, highlighting the leading drug interventions and their respective stages of development. This comprehensive review concludes with a look into the future perspectives, capturing the potential breakthroughs and innovations on the horizon. Through a synthesis of current knowledge and emerging research, this article aims to provide a consolidated resource for clinicians, researchers, and academicians in the realm of Alzheimer's disease.
Collapse
Affiliation(s)
- Monika Sharma
- Faculty of Pharmacy, Department of Pharmacology, Swami Vivekanand Subharti University, Meerut, Uttar Pradesh, India
| | - Pankaj Pal
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, India.
| | - Sukesh Kumar Gupta
- KIET School of Pharmacy, KIET Group of Institutions, Ghaziabad, Uttar Pradesh, India; Department of Ophthalmology, Visual and Anatomical Sciences (OVAS), School of Medicine, Wayne State University, USA.
| |
Collapse
|
4
|
Francisco T, Malafaia D, Melo L, Silva AMS, Albuquerque HMT. Recent Advances in Fluorescent Theranostics for Alzheimer's Disease: A Comprehensive Survey on Design, Synthesis, and Properties. ACS OMEGA 2024; 9:13556-13591. [PMID: 38559945 PMCID: PMC10975685 DOI: 10.1021/acsomega.3c10417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/06/2024] [Accepted: 02/13/2024] [Indexed: 04/04/2024]
Abstract
Alzheimer's disease (AD) is the most common form of neurodegenerative dementia that is rapidly becoming a major health problem, especially in developed countries because of their increasing life expectancy. Two main problems are often associated with the disease: (i) the absence of a widely accessible "gold-standard" for early diagnosis and (ii) lack of effective therapies with disease-modifying effects. The recent success of the monoclonal antibody lecanemab played an important role not only in clarifying a possible druggable pathway but also in spelling the revival of small molecule drug discovery. Unlike bulky biologics, small molecules are structurally less complex, generally cheaper, and compatible with at-home oral consumption, making it feasible for people to start their drug regimen early and stay on it longer. In this sense, small-molecule near-infrared fluorescent theranostics have been gaining more and more attention from the scientific community, as they have the potential to simultaneously provide diagnostic outputs and deliver therapeutic action, paving the way toward personalized medicine in AD patients. They also have the potential to shift the diagnostic "status-quo" from expensive and limited-access PET radiotracers toward inexpensive and handy imaging tools widely available for primary patient screening and preclinical animal studies. Herein, we review the most recent advances in the field of fluorescent theranostics for Alzheimer's disease, detailing their design strategies, synthetic approaches and imaging and therapeutic properties in vitro and in vivo. With this Review, we intend to provide a milestone in the acquired knowledge in the field of AD theranostics, encouraging the future development of properly designed theranostic compounds with improved chances to reach clinical applications.
Collapse
Affiliation(s)
- Telmo
N. Francisco
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus
de Santiago, 3810-193 Aveiro, Portugal
| | - Daniela Malafaia
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus
de Santiago, 3810-193 Aveiro, Portugal
| | - Lúcia Melo
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus
de Santiago, 3810-193 Aveiro, Portugal
| | - Artur M. S. Silva
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus
de Santiago, 3810-193 Aveiro, Portugal
| | - Hélio M. T. Albuquerque
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus
de Santiago, 3810-193 Aveiro, Portugal
| |
Collapse
|
5
|
Zorkina YA, Morozova IO, Abramova OV, Ochneva AG, Gankina OA, Andryushenko AV, Kurmyshev MV, Kostyuk GP, Morozova AY. [Use of modern classification systems for complex diagnostics of Alzheimer's disease]. Zh Nevrol Psikhiatr Im S S Korsakova 2024; 124:121-127. [PMID: 38261294 DOI: 10.17116/jnevro2024124011121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
OBJECTIVE To compare the content of β-amyloid (Aβ) peptides Aβ40, Aβ42, total and threonine phosphorylated 181 tau-protein in cerebrospinal fluid (CSF) of patients with the clinical diagnosis of Alzheimer's disease (AD). MATERIAL AND METHODS The study was performed on 64 patients with a diagnosis of dementia and MMSE scores of 24 or lower. All patients underwent lumbar puncture. Aβ40, Aβ42, Aβ42/40 ratio, total tau, phosphorylated tau at threonine 181 were determined in the CSF using a multiplex assay according to the manufacturer's protocol, the concentration was determined in pkg/ml. RESULTS The preliminary diagnosis of AD was made in 3 patients (5%). As a result of the study of protein content in the CSF, signs of AD were detected in 48 (75%) people. The findings suggest that the diagnosis of AD is made 10-14 times less frequently than it should be according to the World Health Organization data. The discrepancy between clinical diagnosis and laboratory findings is confirmed by our study. CONCLUSION Differences in the therapy of dementias and the development of new drugs targeting specific links in the pathogenesis of different types of dementias require accurate and complete diagnosis of dementias, especially AD, as the most common type of dementia.
Collapse
Affiliation(s)
- Y A Zorkina
- Serbsky National Medical Research Center of Psychiatry and Narcology, Moscow, Russia
- Alexeev Mental-Health Clinic No. 1 of Moscow Healthcare Department, Moscow, Russia
| | - I O Morozova
- Alexeev Mental-Health Clinic No. 1 of Moscow Healthcare Department, Moscow, Russia
| | - O V Abramova
- Serbsky National Medical Research Center of Psychiatry and Narcology, Moscow, Russia
- Alexeev Mental-Health Clinic No. 1 of Moscow Healthcare Department, Moscow, Russia
| | - A G Ochneva
- Serbsky National Medical Research Center of Psychiatry and Narcology, Moscow, Russia
- Alexeev Mental-Health Clinic No. 1 of Moscow Healthcare Department, Moscow, Russia
| | - O A Gankina
- Alexeev Mental-Health Clinic No. 1 of Moscow Healthcare Department, Moscow, Russia
- Russian Medical Academy of Continuous Professional Education, Moscow, Russia
| | - A V Andryushenko
- Alexeev Mental-Health Clinic No. 1 of Moscow Healthcare Department, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | - M V Kurmyshev
- Alexeev Mental-Health Clinic No. 1 of Moscow Healthcare Department, Moscow, Russia
| | - G P Kostyuk
- Alexeev Mental-Health Clinic No. 1 of Moscow Healthcare Department, Moscow, Russia
| | - A Yu Morozova
- Serbsky National Medical Research Center of Psychiatry and Narcology, Moscow, Russia
- Alexeev Mental-Health Clinic No. 1 of Moscow Healthcare Department, Moscow, Russia
| |
Collapse
|
6
|
Bittner N, Funk CSM, Schmidt A, Bermpohl F, Brandl EJ, Algharably EEA, Kreutz R, Riemer TG. Psychiatric Adverse Events of Acetylcholinesterase Inhibitors in Alzheimer's Disease and Parkinson's Dementia: Systematic Review and Meta-Analysis. Drugs Aging 2023; 40:953-964. [PMID: 37682445 PMCID: PMC10600312 DOI: 10.1007/s40266-023-01065-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND The acetylcholinesterase inhibitors (AChEIs) donepezil, galantamine, and rivastigmine are commonly used in the management of various forms of dementia. OBJECTIVES While these drugs are known to induce classic cholinergic adverse events such as diarrhea, their potential to cause psychiatric adverse events has yet to be thoroughly examined. METHODS We sought to determine the risk of psychiatric adverse events associated with the use of AChEIs through a systematic review and meta-analysis of double-blind randomized controlled trials involving patients with Alzheimer's dementia and Parkinson's dementia. RESULTS A total of 48 trials encompassing 22,845 patients were included in our analysis. Anorexia was the most commonly reported psychiatric adverse event, followed by agitation, insomnia, and depression. Individuals exposed to AChEIs had a greater risk of experiencing appetite disorders, insomnia, or depression compared with those who received placebo (anorexia: odds ratio [OR] 2.93, 95% confidence interval [CI] 2.29-3.75; p < 0.00001; decreased appetite: OR 1.93, 95% CI 1.33-2.82; p = 0.0006; insomnia: OR 1.55, 95% CI 1.25-1.93; p < 0.0001; and depression: OR 1.59, 95% CI 1.23-2.06, p = 0.0004). Appetite disorders were also more frequent with high-dose versus low-dose therapy. A subgroup analysis revealed that the risk of insomnia was higher for donepezil than for galantamine. CONCLUSIONS Our findings suggest that AChEI therapy may negatively impact psychological health, and careful monitoring of new psychiatric symptoms is warranted. Lowering the dose may resolve some psychiatric adverse events, as may switching to galantamine in the case of insomnia. CLINICAL TRIAL REGISTRATION The study was pre-registered on PROSPERO (CRD42021258376).
Collapse
Affiliation(s)
- Nadine Bittner
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Cleo S. M. Funk
- Institute of Clinical Pharmacology and Toxicology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany
| | - Alexander Schmidt
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Felix Bermpohl
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Eva J. Brandl
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Engi E. A. Algharably
- Institute of Clinical Pharmacology and Toxicology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany
| | - Reinhold Kreutz
- Institute of Clinical Pharmacology and Toxicology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany
| | - Thomas G. Riemer
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
- Institute of Clinical Pharmacology and Toxicology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany
| |
Collapse
|
7
|
Shahid K, Tamene Y, Mody SP, Sadiq KO, Shivakumar YM, Burra E, Ramphall S. Comparative Study of Safety and Efficacy of Angiotensin-Receptor Blockers and Anti Amyloid-ß Monoclonal Antibodies for the Treatment of Alzheimer's Disease: A Systematic Review. Cureus 2023; 15:e43984. [PMID: 37746412 PMCID: PMC10516255 DOI: 10.7759/cureus.43984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
Amyloid-ß (Aß) plaques and Neurofibrillary tangles are hallmarks of Alzheimer's disease (AD) pathology. Recent advances to find a cure for AD have led to the exploration of Anti-Aß monoclonal antibodies and angiotensin-receptor blockers (ARBs). The antibodies can decrease plaque formation or remove already formed plaques. ARBs increase angiotensin II (AT2) levels and decrease the effect of AT2 on the AT1 receptor (AT1R). This systematic analysis reviews evidence of monoclonal antibodies (Aducanumab, Lecanemab, Donanemab, and Solanezumab) and ARBs in managing AD. An in-depth methodical search was conducted across PubMed, Science Direct, and Mendeley. PRISMA 2020 guidelines were followed for this study. Randomized control trials for antibodies and ARBs and one retrospective cohort study were included. The comparison was made among studies that shared similar measured outcomes. Antibodies were found to be more effective than ARBs, with Aducanumab and Lecanemab being the most effective. ARBs, on the other hand, were found to be the safer choice. Further trials of longer duration and larger sample sizes are needed to explore both groups' long-term safety and efficacy.
Collapse
Affiliation(s)
- Kamran Shahid
- Internal Medicine/Family Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Yonas Tamene
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Shefali P Mody
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Kaiser O Sadiq
- General Surgery, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Yogamba M Shivakumar
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Eshwar Burra
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Shivana Ramphall
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
8
|
Vásquez-Londoño CA, Howes MJR, Costa GM, Arboleda G, Rojas-Cardozo MA. Scutellaria incarnata Vent. root extract and isolated phenylethanoid glycosides are neuroprotective against C 2-ceramide toxicity. JOURNAL OF ETHNOPHARMACOLOGY 2023; 307:116218. [PMID: 36738946 DOI: 10.1016/j.jep.2023.116218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/13/2023] [Accepted: 01/27/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Neuronal death is a central process in neurodegenerative diseases and represents a therapeutic challenge for their prevention and treatment. Scutellaria incarnata Vent. roots are used traditionally in Colombia for central nervous system conditions including those affecting cognitive functions, but their chemistry and neuroprotective action remain to be explored to understand the scientific basis for their medicinal uses. In this study, S. incarnata roots are investigated to assess whether they have neuroprotective effects that could provide some explanation for their traditional use in neurodegenerative diseases. AIM OF THE STUDY To evaluate the neuroprotective effect of S. incarnata roots and its chemical constituents against C2-ceramide-induced cell death in Cath.-a-differentiated (CAD) cells. MATERIALS AND METHODS S. incarnata root ethanol extract was fractionated and compounds were isolated by column chromatography; their structures were elucidated by nuclear magnetic resonance spectroscopy, mass spectrometry and infrared spectroscopy. The cytotoxic and neuroprotective effects against C2-ceramide of S. incarnata root extract, fractions and isolated compounds were assessed in CAD cells. RESULTS S. incarnata root extract and its n-butanol fraction were not cytotoxic but showed neuroprotective effects against C2-ceramide toxicity in CAD cells. The phenylethanoid glycosides incarnatoside (isolated for the first time) and stachysoside C (12.5, 25 and 50 μg/mL) from S. incarnata roots also protected CAD cells against C2-ceramide without inducing cytotoxic effects. CONCLUSION The observed neuroprotective effects of S. incarnata root extract and isolated phenylethanoid glycosides in CAD cells provide an ethnopharmacological basis for the traditional use of this species in Colombia for central nervous system disorders.
Collapse
Affiliation(s)
- Carlos A Vásquez-Londoño
- Universidad Nacional de Colombia-Sede Bogotá, Faculty of Sciences, Department of Pharmacy, GIFFUN, Bogotá, 111321, Colombia.
| | | | - Geison M Costa
- Pontificia Universidad Javeriana, Faculty of Sciences, Department of Pharmacy, Bogotá, 110231, Colombia
| | - Gonzalo Arboleda
- Universidad Nacional de Colombia-Sede Bogotá, Faculty of Medicine, Department of Pathology - Genetic Institute, Bogotá, 111231, Colombia
| | - Maritza A Rojas-Cardozo
- Universidad Nacional de Colombia-Sede Bogotá, Faculty of Sciences, Department of Pharmacy, GIFFUN, Bogotá, 111321, Colombia.
| |
Collapse
|
9
|
Nguyen TH, Wang SL, Nguyen VB. Microorganism-Derived Molecules as Enzyme Inhibitors to Target Alzheimer's Diseases Pathways. Pharmaceuticals (Basel) 2023; 16:ph16040580. [PMID: 37111337 PMCID: PMC10146315 DOI: 10.3390/ph16040580] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. It increases the risk of other serious diseases and causes a huge impact on individuals, families, and socioeconomics. AD is a complex multifactorial disease, and current pharmacological therapies are largely based on the inhibition of enzymes involved in the pathogenesis of AD. Natural enzyme inhibitors are the potential sources for targeting AD treatment and are mainly collected from plants, marine organisms, or microorganisms. In particular, microbial sources have many advantages compared to other sources. While several reviews on AD have been reported, most of these previous reviews focused on presenting and discussing the general theory of AD or overviewing enzyme inhibitors from various sources, such as chemical synthesis, plants, and marine organisms, while only a few reviews regarding microbial sources of enzyme inhibitors against AD are available. Currently, multi-targeted drug investigation is a new trend for the potential treatment of AD. However, there is no review that has comprehensively discussed the various kinds of enzyme inhibitors from the microbial source. This review extensively addresses the above-mentioned aspect and simultaneously updates and provides a more comprehensive view of the enzyme targets involved in the pathogenesis of AD. The emerging trend of using in silico studies to discover drugs concerning AD inhibitors from microorganisms and perspectives for further experimental studies are also covered here.
Collapse
Affiliation(s)
- Thi Hanh Nguyen
- Doctoral Program in Applied Sciences, Tamkang University, New Taipei City 25137, Taiwan
- Department of Chemistry, Tamkang University, New Taipei City 25137, Taiwan
| | - San-Lang Wang
- Department of Chemistry, Tamkang University, New Taipei City 25137, Taiwan
| | - Van Bon Nguyen
- Institute of Biotechnology and Environment, Tay Nguyen University, Buon Ma Thuot 630000, Vietnam
| |
Collapse
|
10
|
Wojtunik-Kulesza K, Rudkowska M, Orzeł-Sajdłowska A. Aducanumab-Hope or Disappointment for Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24054367. [PMID: 36901797 PMCID: PMC10002282 DOI: 10.3390/ijms24054367] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
In June 2021, the world was informed about a new drug for Alzheimer's disease approved by the FDA. Aducanumab (BIIB037, ADU), being a monoclonal antibody IgG1, is the newest AD treatment. The activity of the drug is targeted towards amyloid β, which is considered one of the main causes of Alzheimer's disease. Clinical trials have revealed time- and dose-dependent activity towards Aβ reduction, as well as cognition improvement. Biogen, the company responsible for conducting research and introducing the drug to the market, presents the drug as a solution to cognitive impairment, but its limitations, costs, and side effects are controversial. The framework of the paper focuses on the mechanism of aducanumab's action along with the positive and negative sides of the therapy. The review presents the basis of the amyloid hypothesis that is the cornerstone of therapy, as well as the latest information about aducanumab, its mechanism of action, and the possibility of the use of the drug.
Collapse
Affiliation(s)
- Karolina Wojtunik-Kulesza
- Department of Inorganic Chemistry, Medical University of Lublin, 20-059 Lublin, Poland
- Correspondence:
| | - Monika Rudkowska
- Independent Laboratory of Behavioral Studies, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland
| | | |
Collapse
|
11
|
Porosk L, Härk HH, Bicev RN, Gaidutšik I, Nebogatova J, Armolik EJ, Arukuusk P, da Silva ER, Langel Ü. Aggregation Limiting Cell-Penetrating Peptides Derived from Protein Signal Sequences. Int J Mol Sci 2023; 24:ijms24054277. [PMID: 36901707 PMCID: PMC10002422 DOI: 10.3390/ijms24054277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease (ND) and the leading cause of dementia. It is characterized by non-linear, genetic-driven pathophysiological dynamics with high heterogeneity in the biological alterations and the causes of the disease. One of the hallmarks of the AD is the progression of plaques of aggregated amyloid-β (Aβ) or neurofibrillary tangles of Tau. Currently there is no efficient treatment for the AD. Nevertheless, several breakthroughs in revealing the mechanisms behind progression of the AD have led to the discovery of possible therapeutic targets. Some of these include the reduction in inflammation in the brain, and, although highly debated, limiting of the aggregation of the Aβ. In this work we show that similarly to the Neural cell adhesion molecule 1 (NCAM1) signal sequence, other Aβ interacting protein sequences, especially derived from Transthyretin, can be used successfully to reduce or target the amyloid aggregation/aggregates in vitro. The modified signal peptides with cell-penetrating properties reduce the Aβ aggregation and are predicted to have anti-inflammatory properties. Furthermore, we show that by expressing the Aβ-EGFP fusion protein, we can efficiently assess the potential for reduction in aggregation, and the CPP properties of peptides in mammalian cells.
Collapse
Affiliation(s)
- Ly Porosk
- Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia
- Correspondence:
| | - Heleri Heike Härk
- Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia
| | - Renata Naporano Bicev
- Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil
| | - Ilja Gaidutšik
- Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia
| | | | - Eger-Jasper Armolik
- Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia
| | - Piret Arukuusk
- Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia
| | | | - Ülo Langel
- Institute of Technology, University of Tartu, Nooruse 1, 50411 Tartu, Estonia
- Department Biochemistry and Biophysics, Stockholm University, S.Arrheniusv. 16B, Room C472, 106 91 Stockholm, Sweden
| |
Collapse
|
12
|
Abstract
Although the cause(s) of Alzheimer's disease in the majority of cases remains elusive, it has long been associated with hypertension. In animal models of the disease, hypertension has been shown to exacerbate Alzheimer-like pathology and behavior, while in humans, hypertension during mid-life increases the risk of developing the disease later in life. Unfortunately, once individuals are diagnosed with the disease, there are few therapeutic options available. There is neither an effective symptomatic treatment, one that treats the debilitating cognitive and memory deficits, nor, more importantly, a neuroprotective treatment, one that stops the relentless progression of the pathology. Further, there is no specific preventative treatment that offsets the onset of the disease. A key factor or clue in this quest for an effective preventative and therapeutic treatment may lie in the contribution of hypertension to the disease. In this review, we explore the idea that photobiomodulation, the application of specific wavelengths of light onto body tissues, can reduce the neuropathology and behavioral deficits in Alzheimer's disease by controlling hypertension. We suggest that treatment with photobiomodulation can be an effective preventative and therapeutic option for this neurodegenerative disease.
Collapse
Affiliation(s)
- Audrey Valverde
- Université Grenoble Alpes, Fonds de dotation Clinatec, Grenoble, France
| | - John Mitrofanis
- Université Grenoble Alpes, Fonds de dotation Clinatec, Grenoble, France,
Institute of Ophthalmology, University College London, London, United Kingdom,Correspondence to: John Mitrofanis, E-mail:
| |
Collapse
|
13
|
Jang HY, Oh JM, Kim IW. Drug repurposing using meta-analysis of gene expression in Alzheimer's disease. Front Neurosci 2022; 16:989174. [PMID: 36440278 PMCID: PMC9684643 DOI: 10.3389/fnins.2022.989174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/19/2022] [Indexed: 01/25/2023] Open
Abstract
INTRODUCTION Alzheimer's disease and other forms of dementia are disease that bring an increased global burden. However, the medicine developed to date remains limited. The purpose of this study is to predict drug repositioning candidates using a computational method that integrates gene expression profiles on Alzheimer's disease and compound-induced changes in gene expression levels. METHODS Gene expression data on Alzheimer's disease were obtained from the Gene Expression Omnibus (GEO) and we conducted a meta-analysis of their gene expression levels. The reverse scores of compound-induced gene expressions were computed based on the reversal relationship between disease and drug gene expression profiles. RESULTS Reversal genes and the candidate compounds were identified by the leave-one-out cross-validation procedure. Additionally, the half-maximal inhibitory concentration (IC50) values and the blood-brain barrier (BBB) permeability of candidate compounds were obtained from ChEMBL and PubChem, respectively. CONCLUSION New therapeutic target genes and drug candidates against Alzheimer's disease were identified by means of drug repositioning.
Collapse
Affiliation(s)
- Ha Young Jang
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Jung Mi Oh
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea,College of Pharmacy, Seoul National University, Seoul, South Korea
| | - In-Wha Kim
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea,*Correspondence: In-Wha Kim,
| |
Collapse
|
14
|
Therapeutic Approach to Alzheimer’s Disease: Current Treatments and New Perspectives. Pharmaceutics 2022; 14:pharmaceutics14061117. [PMID: 35745693 PMCID: PMC9228613 DOI: 10.3390/pharmaceutics14061117] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of dementia. The pathophysiology of this disease is characterized by the accumulation of amyloid-β, leading to the formation of senile plaques, and by the intracellular presence of neurofibrillary tangles based on hyperphosphorylated tau protein. In the therapeutic approach to AD, we can identify three important fronts: the approved drugs currently available for the treatment of the disease, which include aducanumab, donepezil, galantamine, rivastigmine, memantine, and a combination of memantine and donepezil; therapies under investigation that work mainly on Aβ pathology and tau pathology, and which include γ-secretase inhibitors, β-secretase inhibitors, α-secretase modulators, aggregation inhibitors, metal interfering drugs, drugs that enhance Aβ clearance, inhibitors of tau protein hyperphosphorylation, tau protein aggregation inhibitors, and drugs that promote the clearance of tau, and finally, other alternative therapies designed to improve lifestyle, thus contributing to the prevention of the disease. Therefore, the aim of this review was to analyze and describe current treatments and possible future alternatives in the therapeutic approach to AD.
Collapse
|
15
|
Piccialli I, Tedeschi V, Caputo L, D’Errico S, Ciccone R, De Feo V, Secondo A, Pannaccione A. Exploring the Therapeutic Potential of Phytochemicals in Alzheimer’s Disease: Focus on Polyphenols and Monoterpenes. Front Pharmacol 2022; 13:876614. [PMID: 35600880 PMCID: PMC9114803 DOI: 10.3389/fphar.2022.876614] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/11/2022] [Indexed: 12/21/2022] Open
Abstract
Alzheimer’s disease (AD) is a chronic, complex neurodegenerative disorder mainly characterized by the irreversible loss of memory and cognitive functions. Different hypotheses have been proposed thus far to explain the etiology of this devastating disorder, including those centered on the Amyloid-β (Aβ) peptide aggregation, Tau hyperphosphorylation, neuroinflammation and oxidative stress. Nonetheless, the therapeutic strategies conceived thus far to treat AD neurodegeneration have proven unsuccessful, probably due to the use of single-target drugs unable to arrest the progressive deterioration of brain functions. For this reason, the theoretical description of the AD etiology has recently switched from over-emphasizing a single deleterious process to considering AD neurodegeneration as the result of different pathogenic mechanisms and their interplay. Moreover, much relevance has recently been conferred to several comorbidities inducing insulin resistance and brain energy hypometabolism, including diabetes and obesity. As consequence, much interest is currently accorded in AD treatment to a multi-target approach interfering with different pathways at the same time, and to life-style interventions aimed at preventing the modifiable risk-factors strictly associated with aging. In this context, phytochemical compounds are emerging as an enormous source to draw on in the search for multi-target agents completing or assisting the traditional pharmacological medicine. Intriguingly, many plant-derived compounds have proven their efficacy in counteracting several pathogenic processes such as the Aβ aggregation, neuroinflammation, oxidative stress and insulin resistance. Many strategies have also been conceived to overcome the limitations of some promising phytochemicals related to their poor pharmacokinetic profiles, including nanotechnology and synthetic routes. Considering the emerging therapeutic potential of natural medicine, the aim of the present review is therefore to highlight the most promising phytochemical compounds belonging to two major classes, polyphenols and monoterpenes, and to report the main findings about their mechanisms of action relating to the AD pathogenesis.
Collapse
Affiliation(s)
- Ilaria Piccialli
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, University of Naples “Federico II”, Naples, Italy
| | - Valentina Tedeschi
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, University of Naples “Federico II”, Naples, Italy
| | - Lucia Caputo
- Department of Pharmacy, University of Salerno, Salerno, Italy
| | - Stefano D’Errico
- Department of Pharmacy, University of Naples “Federico II”, Naples, Italy
| | - Roselia Ciccone
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, University of Naples “Federico II”, Naples, Italy
| | - Vincenzo De Feo
- Department of Pharmacy, University of Salerno, Salerno, Italy
| | - Agnese Secondo
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, University of Naples “Federico II”, Naples, Italy
| | - Anna Pannaccione
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, University of Naples “Federico II”, Naples, Italy
- *Correspondence: Anna Pannaccione,
| |
Collapse
|
16
|
Dorababu A. Promising heterocycle-based scaffolds in recent (2019-2021) anti-Alzheimer's drug design and discovery. Eur J Pharmacol 2022; 920:174847. [PMID: 35218718 DOI: 10.1016/j.ejphar.2022.174847] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/06/2022] [Accepted: 02/18/2022] [Indexed: 12/28/2022]
Abstract
Alzheimer's disease (AD) is one of the neurodegenerative diseases that led to morbidity and mortality world-wide. It is a complex disease whose etiology is not completely known that leads to difficulty in prevent or cure of the AD. Also, there are only few approved drugs for AD treatment. Apart from deaths due to AD, expenditure of treatment and care of AD patients is higher than that of treatment of HIV and cancer diseases combined. Hence, it leads to an economic burden also. Although research is being carried out on designing drugs for AD, most of them have ended up in poor inhibitors with high toxicity. Hence, researchers should shoulder a great responsibility of discovery of efficient drugs for AD treatment. In the field of drug discovery, heterocycles played an important role. Also, most of the heterocyclic scaffolds have been used in design of potent anti-AD agents. In view of this, heterocyclic molecules reported recently are compiled and evaluated comprehensively. Especially, the molecules which exhibited pronounced activity are emphasized and described with respect to structure-activity relationship (SAR) in brief.
Collapse
Affiliation(s)
- Atukuri Dorababu
- SRMPP Government First Grade College, Huvinahadagali, 583219, India.
| |
Collapse
|
17
|
Small molecules targeting γ-secretase and their potential biological applications. Eur J Med Chem 2022; 232:114169. [DOI: 10.1016/j.ejmech.2022.114169] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 01/30/2022] [Accepted: 01/30/2022] [Indexed: 12/14/2022]
|
18
|
Tripathi AS, Bansod P, Swathi KP. Activation of 5-HT 1b/d receptor restores the cognitive function by reducing glutamate release, deposition of β-amyloid and TLR-4 pathway in the brain of scopolamine-induced dementia in rat. J Pharm Pharmacol 2021; 73:1592-1598. [PMID: 34244776 DOI: 10.1093/jpp/rgab095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 06/07/2021] [Indexed: 11/13/2022]
Abstract
OBJECTIVES This study evaluates the effect of 5-HT 1b/d agonist on cognitive function in scopolamine (SPN)-induced dementia in the rat. METHODS Dementia was induced by administration of SPN 2 mg/kg/day, intraperitoneally, for a duration of 21 days. The effect of zolmitriptan (ZMT) 30 mg/kg, intraperitoneally, was observed on cognitive function, and the parameters of oxidative stress like malondialdehyde (MDA) level, nitric oxide (NO), superoxide dismutase (SOD) and glutathione peroxidase (GPX) were estimated at the end. Histopathology study of brain tissue was performed for the determination of β-amyloid peptide, and qRT-PCR was used to determine the mRNA expression of Toll-like receptor 4 (TLR-4), IL-17 and β-amyloid. KEY FINDINGS Data of the study suggested that treatment with ZMT alone and in combination with DMP (dextromethorphan) significantly (P < 0.01) decreases the escape latency in conditioned avoidance response (CAR) and transfer latency in elevated plus maze (EPM) as compared with negative control group. Moreover, the result of Morris water maze (MWM) shows an increase in retention time and a decrease in escape latency in ZMT alone and in combination with DMP-treated group of SPN-induced dementia than in the negative control group. There was a significant decrease in MDA and NO and increase in SOD and GPX in the brain tissues of ZMT and ZMT + DMP-treated group than negative control group. Histopathology study also suggested that the concentration of Aβ peptide decreases in the brain tissues in ZMT and ZMT + DMP-treated group than the negative control group. Moreover, ZMT treatment ameliorates the altered mRNA expression of TLR-4 and IL-17 in the brain tissue of SPN-induced dementia rat. CONCLUSIONS In conclusion, ZMT restores the cognitive functions and impaired memory in SPN-induced dementia in the rat by decreasing oxidative stress and Aβ peptide in the brain tissue of rat.
Collapse
Affiliation(s)
- Alok Shiomurti Tripathi
- Amity Institute of Pharmacy, Amity University, Sector 125, Noida, Uttar Pradesh, India
- Department of Pharmacology, P. Wadhwani College of Pharmacy, Yavatmal, Maharashtra, India
| | - Prajakta Bansod
- Department of Pharmacology, P. Wadhwani College of Pharmacy, Yavatmal, Maharashtra, India
| | - K P Swathi
- Department of Pharmacology, College of Pharmacy, Kannur Medical College, Kannur, Kerala, India
| |
Collapse
|
19
|
Jeremic D, Jiménez-Díaz L, Navarro-López JD. Past, present and future of therapeutic strategies against amyloid-β peptides in Alzheimer's disease: a systematic review. Ageing Res Rev 2021; 72:101496. [PMID: 34687956 DOI: 10.1016/j.arr.2021.101496] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/30/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease in ageing, affecting around 46 million people worldwide but few treatments are currently available. The etiology of AD is still puzzling, and new drugs development and clinical trials have high failure rates. Urgent outline of an integral (multi-target) and effective treatment of AD is needed. Accumulation of amyloid-β (Aβ) peptides is considered one of the fundamental neuropathological pillars of the disease, and its dyshomeostasis has shown a crucial role in AD onset. Therefore, many amyloid-targeted therapies have been investigated. Here, we will systematically review recent (from 2014) investigational, follow-up and review studies focused on anti-amyloid strategies to summarize and analyze their current clinical potential. Combination of anti-Aβ therapies with new developing early detection biomarkers and other therapeutic agents acting on early functional AD changes will be highlighted in this review. Near-term approval seems likely for several drugs acting against Aβ, with recent FDA approval of a monoclonal anti-Aβ oligomers antibody -aducanumab- raising hopes and controversies. We conclude that, development of oligomer-epitope specific Aβ treatment and implementation of multiple improved biomarkers and risk prediction methods allowing early detection, together with therapies acting on other factors such as hyperexcitability in early AD, could be the key to slowing this global pandemic.
Collapse
|
20
|
Gupta GL, Samant NP. Current druggable targets for therapeutic control of Alzheimer's disease. Contemp Clin Trials 2021; 109:106549. [PMID: 34464763 DOI: 10.1016/j.cct.2021.106549] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative brain disorder that has an increasingly large burden on health and social care systems. The pathophysiology involves the accumulation of extracellular amyloid-beta plaques (Aβ) and intracellular neurofibrillary tangles contributing to neuronal death and leading to cognition impairment. However, its cause remains poorly understood, and there is no cure for AD despite extensive research and billions of dollars spent over decades. Currently, there are only four US Food and Drug Administration (FDA) approved drugs and one combination therapy available in the market for the symptomatic relief of AD. Since 2003, no new drug has been approved by the FDA for the treatment of AD. Researchers continue to explore new treatments and therapeutic strategies to treat AD. The need for novel discoveries on therapeutic targets and the development of new therapeutic approaches is imminent when considering the current expectations regarding the increased number of AD cases each year and the huge financial cost amounted to healthcare. This review focused on the current status of drugs in the clinical pipeline targeting β-amyloid, tau phosphorylation, or neurotransmitter dysfunction for therapeutic control of Alzheimer's disease.
Collapse
Affiliation(s)
- Girdhari Lal Gupta
- School of Pharmacy & Technology Management, SVKM'S NMIMS, Shirpur Campus, Shirpur 425 405, Maharashtra, India; Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400 056, Maharashtra, India.
| | - Nikita Patil Samant
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400 056, Maharashtra, India
| |
Collapse
|
21
|
Sahoo BR, Panda PK, Liang W, Tang WJ, Ahuja R, Ramamoorthy A. Degradation of Alzheimer's Amyloid-β by a Catalytically Inactive Insulin-Degrading Enzyme. J Mol Biol 2021; 433:166993. [PMID: 33865867 PMCID: PMC8169600 DOI: 10.1016/j.jmb.2021.166993] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 03/24/2021] [Accepted: 04/06/2021] [Indexed: 02/06/2023]
Abstract
It is known that insulin-degrading-enzyme (IDE) plays a crucial role in the clearance of Alzheimer's amyloid-β (Aβ). The cysteine-free IDE mutant (cf-E111Q-IDE) is catalytically inactive against insulin, but its effect on Aβ degradation is unknown that would help in the allosteric modulation of the enzyme activity. Herein, the degradation of Aβ(1-40) by cf-E111Q-IDE via a non-chaperone mechanism is demonstrated by NMR and LC-MS, and the aggregation of fragmented peptides is characterized using fluorescence and electron microscopy. cf-E111Q-IDE presented a reduced effect on the aggregation kinetics of Aβ(1-40) when compared with the wild-type IDE. Whereas LC-MS and diffusion ordered NMR spectroscopy revealed the generation of Aβ fragments by both wild-type and cf-E111Q-IDE. The aggregation propensities and the difference in the morphological phenotype of the full-length Aβ(1-40) and its fragments are explained using multi-microseconds molecular dynamics simulations. Notably, our results reveal that zinc binding to Aβ(1-40) inactivates cf-E111Q-IDE's catalytic function, whereas zinc removal restores its function as evidenced from high-speed AFM, electron microscopy, chromatography, and NMR results. These findings emphasize the catalytic role of cf-E111Q-IDE on Aβ degradation and urge the development of zinc chelators as an alternative therapeutic strategy that switches on/off IDE's function.
Collapse
Affiliation(s)
- Bikash R Sahoo
- Biophysics, Department of Chemistry, Macromolecular Engineering and Science, and Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Pritam Kumar Panda
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-75120 Uppsala, Sweden
| | - Wenguang Liang
- Ben-May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Wei-Jen Tang
- Ben-May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Rajeev Ahuja
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-75120 Uppsala, Sweden; Applied Materials Physics, Department of Materials Science and Engineering, Royal Institute of Technology (KTH) SE-10044 Stockholm, Sweden
| | - Ayyalusamy Ramamoorthy
- Biophysics, Department of Chemistry, Macromolecular Engineering and Science, and Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
22
|
Cyclodextrin Monomers and Polymers for Drug Activity Enhancement. Polymers (Basel) 2021; 13:polym13111684. [PMID: 34064190 PMCID: PMC8196804 DOI: 10.3390/polym13111684] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/13/2022] Open
Abstract
Cyclodextrins (CDs) and cyclodextrin (CD)-based polymers are well-known complexing agents. One of their distinctive features is to increase the quantity of a drug in a solution or improve its delivery. However, in certain instances, the activity of the solutions is increased not only due to the increase of the drug dose but also due to the drug complexation. Based on numerous studies reviewed, the drug appeared more active in a complex form. This review aims to summarize the performance of CDs and CD-based polymers as activity enhancers. Accordingly, the review is divided into two parts, i.e., the effect of CDs as active drugs and as enhancers in antimicrobials, antivirals, cardiovascular diseases, cancer, neuroprotective agents, and antioxidants.
Collapse
|
23
|
Pei YA, Davies J, Zhang M, Zhang HT. The Role of Synaptic Dysfunction in Alzheimer's Disease. J Alzheimers Dis 2021; 76:49-62. [PMID: 32417776 DOI: 10.3233/jad-191334] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Deemed as incurable, Alzheimer's disease (AD) research is becoming less convoluted as our understanding of its pathology increases. With current treatments focusing on merely mitigating the symptoms of AD, there have been many attempts to find a molecular culprit to serve as the single underlying cause and therapeutic target for clinical applications to approach the disease from its roots. Indeed, over the course of decades, the endless search for a singular target culprit in AD has uncovered a cascade of pathological defects, adding on to each other throughout the progression of the disease. The developmental patterns of amyloid-β (Aβ) oligomers have been studied as a means to discover the complex molecular interplay between various immune responses, genetic mutations, pathway disturbances, and regulating factors that disturb synapse homeostasis before disease manifestation. This new understanding has shifted the underlying goal of the research community from merely removing Aβ oligomers to finding methods that can predict high risk individuals and resorting to cocktail-drug treatments in an attempt to regulate multiple pathways that cumulatively result in the debilitating symptoms of the disease. By utilizing various assays from immuno-targeting to molecular biomarkers, we then interfere in the molecular cascades in an endeavor to avoid synapse dysfunction before disease maturity. Here, we review the current literature supporting the importance of synapses in AD, our current understanding of the molecular interactions leading up to clinical diagnoses, and the techniques used in targeted therapies.
Collapse
Affiliation(s)
- Yixuan Amy Pei
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Julie Davies
- Department of Physiology, Anatomy & Genetics, Oxford University, Oxford, UK
| | - Melanie Zhang
- Department of Neurobiology, Northwestern University Feinberg School of Medicine, Evanston, IL, USA
| | - Han-Ting Zhang
- Departments of Neuroscience and Behavioral Medicine & Psychiatry, Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, WV, USA
| |
Collapse
|
24
|
|
25
|
β-Amyloid Orchestrates Factor XII and Platelet Activation Leading to Endothelial Dysfunction and Abnormal Fibrinolysis in Alzheimer Disease. Alzheimer Dis Assoc Disord 2021; 35:91-97. [PMID: 33629978 DOI: 10.1097/wad.0000000000000420] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 09/18/2020] [Indexed: 11/25/2022]
Abstract
Alzheimer disease (AD) is the most common form of dementia in humans. However, to date, the cause of sporadic AD (SAD), which is the most frequent form, is still unknown. Although it has not been possible to determine the origin of this disease, the amyloid hypothesis is one of the most accepted to explain the etiology of AD. This hypothesis proposes that the pathogenesis of AD is derived from the toxic effect produced by the amyloid-β (Aβ) peptide in the brain parenchyma, but it does not make clear how Aβ is capable of producing such damage. Furthermore, it has been observed that SAD is accompanied by disruptions in the vascular system, such as damage to the blood-brain barrier. This facilitates the transfer of some systemic proteins, such as fibrinogen, to the brain parenchyma, where Aβ is abundant. Therefore, this Aβ interacts with fibrinogen, which favors the formation of clots resistant to fibrinolysis, inducing a risk of thrombosis and neuroinflammation. Notably, Aβ is not only of neuronal origin; platelets also contribute to high Aβ production in the circulation. The Aβ present in circulation favors the activation of coagulation factor XII, which leads to the generation of thrombin and bradykinin. In addition to Aβ-induced platelet activation, all these events favor the development of inflammatory processes that cause damage to the brain vasculature. This damage represents the beginning of the toxic effects of Aβ, which supports the amyloid hypothesis. This review addresses the relationship between alterations in the vascular and hemostatic systems caused by Aβ and how both alterations contribute to the progression of SAD.
Collapse
|
26
|
The Neurovascular Unit Dysfunction in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22042022. [PMID: 33670754 PMCID: PMC7922832 DOI: 10.3390/ijms22042022] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/06/2021] [Accepted: 01/11/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disease worldwide. Histopathologically, AD presents with two hallmarks: neurofibrillary tangles (NFTs), and aggregates of amyloid β peptide (Aβ) both in the brain parenchyma as neuritic plaques, and around blood vessels as cerebral amyloid angiopathy (CAA). According to the vascular hypothesis of AD, vascular risk factors can result in dysregulation of the neurovascular unit (NVU) and hypoxia. Hypoxia may reduce Aβ clearance from the brain and increase its production, leading to both parenchymal and vascular accumulation of Aβ. An increase in Aβ amplifies neuronal dysfunction, NFT formation, and accelerates neurodegeneration, resulting in dementia. In recent decades, therapeutic approaches have attempted to decrease the levels of abnormal Aβ or tau levels in the AD brain. However, several of these approaches have either been associated with an inappropriate immune response triggering inflammation, or have failed to improve cognition. Here, we review the pathogenesis and potential therapeutic targets associated with dysfunction of the NVU in AD.
Collapse
|
27
|
You YX, Shahar S, Rajab NF, Haron H, Yahya HM, Mohamad M, Din NC, Maskat MY. Effects of 12 Weeks Cosmos caudatus Supplement among Older Adults with Mild Cognitive Impairment: A Randomized, Double-Blind and Placebo-Controlled Trial. Nutrients 2021; 13:nu13020434. [PMID: 33572715 PMCID: PMC7912368 DOI: 10.3390/nu13020434] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 01/21/2021] [Accepted: 01/24/2021] [Indexed: 01/10/2023] Open
Abstract
Cosmos caudatus (CC) contains high flavonoids and might be beneficial in neuroprotection. It has the potential to prevent neurodegenerative diseases. Therefore, we aimed to investigate the effects of 12 weeks of Cosmos caudatus supplement on cognitive function, mood status, blood biochemical profiles and biomarkers among older adults with mild cognitive impairment (MCI) through a double-blind, placebo-controlled trial. The subjects were randomized into CC supplement (n = 24) and placebo group (n = 24). Each of them consumed one capsule of CC supplement (250 mg of CC/capsule) or placebo (500 mg maltodextrin/capsule) twice daily for 12 weeks. Cognitive function and mood status were assessed at baseline, 6th week, and 12th week using validated neuropsychological tests. Blood biochemical profiles and biomarkers were measured at baseline and 12th week. Two-way mixed analysis of variance (ANOVA) analysis showed significant improvements in mini mental state examination (MMSE) (partial η2 = 0.150, p = 0.049), tension (partial η2 = 0.191, p = 0.018), total mood disturbance (partial η2 = 0.171, p = 0.028) and malondialdehyde (MDA) (partial η2 = 0.097, p = 0.047) following CC supplementation. In conclusion, 12 weeks CC supplementation potentially improved global cognition, tension, total mood disturbance, and oxidative stress among older adults with MCI. Larger sample size and longer period of intervention with incorporation of metabolomic approach should be conducted to further investigate the underlying mechanism of CC supplementation in neuroprotection.
Collapse
Affiliation(s)
- Yee Xing You
- Dietetics Programme and Centre for Healthy Aging and Wellness (H-Care), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Suzana Shahar
- Dietetics Programme and Centre for Healthy Aging and Wellness (H-Care), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
- Correspondence: ; Tel.: +60-3-9289-7651
| | - Nor Fadilah Rajab
- Biomedical Science Programme and Centre for Healthy Aging and Wellness (H-Care), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Hasnah Haron
- Nutritional Sciences Programme and Centre for Healthy Aging and Wellness (H-Care), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia; (H.H.); (H.M.Y.)
| | - Hanis Mastura Yahya
- Nutritional Sciences Programme and Centre for Healthy Aging and Wellness (H-Care), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia; (H.H.); (H.M.Y.)
| | - Mazlyfarina Mohamad
- Diagnostic Imaging and Radiotherapy Programme and Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Normah Che Din
- Health Psychology Programme, Centre of Rehabilitation and Special Needs, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Mohamad Yusof Maskat
- Department of Food Sciences, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, UKM, Bangi 43600, Malaysia;
| |
Collapse
|
28
|
Campora M, Francesconi V, Schenone S, Tasso B, Tonelli M. Journey on Naphthoquinone and Anthraquinone Derivatives: New Insights in Alzheimer's Disease. Pharmaceuticals (Basel) 2021; 14:33. [PMID: 33466332 PMCID: PMC7824805 DOI: 10.3390/ph14010033] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/27/2020] [Accepted: 12/30/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that is characterized by memory loss, cognitive impairment, and functional decline leading to dementia and death. AD imposes neuronal death by the intricate interplay of different neurochemical factors, which continue to inspire the medicinal chemist as molecular targets for the development of new agents for the treatment of AD with diverse mechanisms of action, but also depict a more complex AD scenario. Within the wide variety of reported molecules, this review summarizes and offers a global overview of recent advancements on naphthoquinone (NQ) and anthraquinone (AQ) derivatives whose more relevant chemical features and structure-activity relationship studies will be discussed with a view to providing the perspective for the design of viable drugs for the treatment of AD. In particular, cholinesterases (ChEs), β-amyloid (Aβ) and tau proteins have been identified as key targets of these classes of compounds, where the NQ or AQ scaffold may contribute to the biological effect against AD as main unit or significant substructure. The multitarget directed ligand (MTDL) strategy will be described, as a chance for these molecules to exhibit significant potential on the road to therapeutics for AD.
Collapse
Affiliation(s)
| | | | | | | | - Michele Tonelli
- Dipartimento di Farmacia, Università degli Studi di Genova, Viale Benedetto XV, 3, 16132 Genova, Italy; (M.C.); (V.F.); (S.S.); (B.T.)
| |
Collapse
|
29
|
Lieblein T, Zangl R, Martin J, Hoffmann J, Hutchison MJ, Stark T, Stirnal E, Schrader T, Schwalbe H, Morgner N. Structural rearrangement of amyloid-β upon inhibitor binding suppresses formation of Alzheimer's disease related oligomers. eLife 2020; 9:59306. [PMID: 33095161 PMCID: PMC7682991 DOI: 10.7554/elife.59306] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/22/2020] [Indexed: 01/24/2023] Open
Abstract
The formation of oligomers of the amyloid-β peptide plays a key role in the onset of Alzheimer's disease. We describe herein the investigation of disease-relevant small amyloid-β oligomers by mass spectrometry and ion mobility spectrometry, revealing functionally relevant structural attributes. In particular, we can show that amyloid-β oligomers develop in two distinct arrangements leading to either neurotoxic oligomers and fibrils or non-toxic amorphous aggregates. Comprehending the key-attributes responsible for those pathways on a molecular level is a pre-requisite to specifically target the peptide's tertiary structure with the aim to promote the emergence of non-toxic aggregates. Here, we show for two fibril inhibiting ligands, an ionic molecular tweezer and a hydrophobic peptide that despite their different interaction mechanisms, the suppression of the fibril pathway can be deduced from the disappearance of the corresponding structure of the first amyloid-β oligomers.
Collapse
Affiliation(s)
- Tobias Lieblein
- JW Goethe-University, Institute of Physical and Theoretical Chemistry, Frankfurt, Germany
| | - Rene Zangl
- JW Goethe-University, Institute of Physical and Theoretical Chemistry, Frankfurt, Germany
| | - Janosch Martin
- JW Goethe-University, Institute of Physical and Theoretical Chemistry, Frankfurt, Germany
| | - Jan Hoffmann
- JW Goethe-University, Institute of Physical and Theoretical Chemistry, Frankfurt, Germany
| | - Marie J Hutchison
- JW Goethe-University, Institute for Organic Chemistry and Chemical Biology and Center for Biomolecular Magnetic Resonance, Frankfurt am Main, Germany
| | - Tina Stark
- JW Goethe-University, Institute for Organic Chemistry and Chemical Biology and Center for Biomolecular Magnetic Resonance, Frankfurt am Main, Germany
| | - Elke Stirnal
- JW Goethe-University, Institute for Organic Chemistry and Chemical Biology and Center for Biomolecular Magnetic Resonance, Frankfurt am Main, Germany
| | - Thomas Schrader
- University of Duisburg-Essen, Institute of Organic Chemistry, Essen, Germany
| | - Harald Schwalbe
- JW Goethe-University, Institute for Organic Chemistry and Chemical Biology and Center for Biomolecular Magnetic Resonance, Frankfurt am Main, Germany
| | - Nina Morgner
- JW Goethe-University, Institute of Physical and Theoretical Chemistry, Frankfurt, Germany
| |
Collapse
|
30
|
Matencio A, Caldera F, Cecone C, López-Nicolás JM, Trotta F. Cyclic Oligosaccharides as Active Drugs, an Updated Review. Pharmaceuticals (Basel) 2020; 13:E281. [PMID: 33003610 PMCID: PMC7601923 DOI: 10.3390/ph13100281] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 09/26/2020] [Accepted: 09/27/2020] [Indexed: 12/13/2022] Open
Abstract
There have been many reviews of the cyclic oligosaccharide cyclodextrin (CD) and CD-based materials used for drug delivery, but the capacity of CDs to complex different agents and their own intrinsic properties suggest they might also be considered for use as active drugs, not only as carriers. The aim of this review is to summarize the direct use of CDs as drugs, without using its complexing potential with other substances. The direct application of another oligosaccharide called cyclic nigerosyl-1,6-nigerose (CNN) is also described. The review is divided into lipid-related diseases, aggregation diseases, antiviral and antiparasitic activities, anti-anesthetic agent, function in diet, removal of organic toxins, CDs and collagen, cell differentiation, and finally, their use in contact lenses in which no drug other than CDs are involved. In the case of CNN, its application as a dietary supplement and immunological modulator is explained. Finally, a critical structure-activity explanation is provided.
Collapse
Affiliation(s)
- Adrián Matencio
- Dipartimento di Chimica, Università di Torino, via P. Giuria 7, 10125 Torino, Italy; (F.C.); (C.C.); (F.T.)
| | - Fabrizio Caldera
- Dipartimento di Chimica, Università di Torino, via P. Giuria 7, 10125 Torino, Italy; (F.C.); (C.C.); (F.T.)
| | - Claudio Cecone
- Dipartimento di Chimica, Università di Torino, via P. Giuria 7, 10125 Torino, Italy; (F.C.); (C.C.); (F.T.)
| | - José Manuel López-Nicolás
- Departamento de Bioquímica y Biología Molecular A, Unidad Docente de Biología, Facultad de Veterinaria, Regional Campus of International Excellence “Campus Mare Nostrum”, Universidad de Murcia, 30100 Espinardo, Murcia, Spain;
| | - Francesco Trotta
- Dipartimento di Chimica, Università di Torino, via P. Giuria 7, 10125 Torino, Italy; (F.C.); (C.C.); (F.T.)
| |
Collapse
|
31
|
Pinheiro L, Faustino C. Therapeutic Strategies Targeting Amyloid-β in Alzheimer's Disease. Curr Alzheimer Res 2020; 16:418-452. [PMID: 30907320 DOI: 10.2174/1567205016666190321163438] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/16/2019] [Accepted: 03/17/2019] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder linked to protein misfolding and aggregation. AD is pathologically characterized by senile plaques formed by extracellular Amyloid-β (Aβ) peptide and Intracellular Neurofibrillary Tangles (NFT) formed by hyperphosphorylated tau protein. Extensive synaptic loss and neuronal degeneration are responsible for memory impairment, cognitive decline and behavioral dysfunctions typical of AD. Amyloidosis has been implicated in the depression of acetylcholine synthesis and release, overactivation of N-methyl-D-aspartate (NMDA) receptors and increased intracellular calcium levels that result in excitotoxic neuronal degeneration. Current drugs used in AD treatment are either cholinesterase inhibitors or NMDA receptor antagonists; however, they provide only symptomatic relief and do not alter the progression of the disease. Aβ is the product of Amyloid Precursor Protein (APP) processing after successive cleavage by β- and γ-secretases while APP proteolysis by α-secretase results in non-amyloidogenic products. According to the amyloid cascade hypothesis, Aβ dyshomeostasis results in the accumulation and aggregation of Aβ into soluble oligomers and insoluble fibrils. The former are synaptotoxic and can induce tau hyperphosphorylation while the latter deposit in senile plaques and elicit proinflammatory responses, contributing to oxidative stress, neuronal degeneration and neuroinflammation. Aβ-protein-targeted therapeutic strategies are thus a promising disease-modifying approach for the treatment and prevention of AD. This review summarizes recent findings on Aβ-protein targeted AD drugs, including β-secretase inhibitors, γ-secretase inhibitors and modulators, α-secretase activators, direct inhibitors of Aβ aggregation and immunotherapy targeting Aβ, focusing mainly on those currently under clinical trials.
Collapse
Affiliation(s)
- Lídia Pinheiro
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto 1649-003 Lisboa, Portugal
| | - Célia Faustino
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto 1649-003 Lisboa, Portugal
| |
Collapse
|
32
|
Frost CV, Zacharias M. From monomer to fibril: Abeta-amyloid binding to Aducanumab antibody studied by molecular dynamics simulation. Proteins 2020; 88:1592-1606. [PMID: 32666627 DOI: 10.1002/prot.25978] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/04/2020] [Accepted: 05/13/2020] [Indexed: 12/27/2022]
Abstract
Alzheimer's disease is one of the most common causes of dementia. It is believed that the aggregation of short Aβ-peptides to form oligomeric and protofibrillar amyloid assemblies plays a central role for disease-relevant neurotoxicity. In recent years, passive immunotherapy has been introduced as a potential treatment strategy with anti-amyloid antibodies binding to Aβ-amyloids and inducing their subsequent degradation by the immune system. Although so far mostly unsuccessful in clinical studies, the high-dosed application of the monoclonal antibody Aducanumab has shown therapeutic potential that might be attributed to its much greater affinity to Aβ-aggregates vs monomeric Aβ-peptides. In order to better understand how Aducanumab interacts with aggregated Aβ-forms compared to monomers, we have generated structural model complexes based on the known structure of Aducanumab in complex with an Aβ2 - 7 -eptitope. Structural models of Aducanumab bound to full-sequence Aβ1 - 40 -monomers, oligomers, protofilaments and mature fibrils were generated and investigated using extensive molecular dynamics simulations to characterize the flexibility and possible additional interactions. Indeed, an aggregate-specific N-terminal binding motif was found in case of Aducanumab binding to oligomers, protofilaments and fibrils that is located next to but not overlapping with the epitope binding site found in the crystal structure with Aβ2 - 7 . Analysis of binding energetics indicates that this motif binds weaker than the epitope but likely contributes to Aducanumab's preference for aggregated Aβ-species. The predicted aggregate-specific binding motif could potentially serve as a basis to reengineer Aducanumab for further enhanced preference to bind Aβ-aggregates vs monomers.
Collapse
Affiliation(s)
- Christina V Frost
- Physics Department T38, Technical University of Munich, Garching, Germany
| | - Martin Zacharias
- Physics Department T38, Technical University of Munich, Garching, Germany
| |
Collapse
|
33
|
Neshan M, Campbell A, Malakouti SK, Zareii M, Ahangari G. Gene expression of serotonergic markers in peripheral blood mononuclear cells of patients with late-onset Alzheimer's disease. Heliyon 2020; 6:e04716. [PMID: 32904297 PMCID: PMC7452509 DOI: 10.1016/j.heliyon.2020.e04716] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 06/06/2020] [Accepted: 08/11/2020] [Indexed: 01/14/2023] Open
Abstract
Serotonin or 5-hydroxytryptamine (5-HT) is primarily involved in the regulation of learning and memory. Pathological changes in metabolism or functional imbalance of 5-HT has been associated with Alzheimer's disease (AD). The hypothesis tested is that in peripheral blood, markers of the serotonergic pathway can be used as a diagnostic tool for AD. The current study measured the relative expression of 5-HT receptors (5-HTR2A and 5-HTR3A) as well as the 5-HT catalytic enzyme, Monoamine oxidase A (MAO-A) mRNA in Peripheral Blood Mononuclear Cells (PBMCs) of patients with late-onset Alzheimer's disease (LOAD) and age-matched controls. 5-HTR2A, 5-HTR3A, and MAO-A mRNA expressions were examined in PBMCs of 30 patients with LOAD and 30 control individuals. Real-time quantitative PCR was used to measure mRNA expression. The dementia status of patients in this study was assessed using a Mini-Mental State Examination (MMSE). Mean data of relative mRNA expression of 5-HTR2A, 5-HTR3A and MAO-A were significantly lower in PBMCs of patients with LOAD compared with controls. Based on the down-regulation of serotonergic markers in PBMCs, our findings may be another claim to the systemic nature of LOAD. The role of peripheral serotonergic downregulation, in the pathogenesis of AD, needs to be further studied. Given the extremely convenient access to PBMCs, these molecular events may represent more complete dimensions of AD neuropathophysiology or possibly lead to a new direction in studies focused on blood-based markers.
Collapse
Affiliation(s)
- Masoud Neshan
- Department of Medical Genetics, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Arezoo Campbell
- Department of Pharmaceutical Sciences, Western University of Health Sciences, California, USA
| | - Seyed Kazem Malakouti
- Mental Health Research Center, Tehran Institute of Psychiatry–School of Behavioral Sciences and Mental Health, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Zareii
- Mental Health Research Center, Tehran Institute of Psychiatry–School of Behavioral Sciences and Mental Health, Iran University of Medical Sciences, Tehran, Iran
| | - Ghasem Ahangari
- Department of Medical Genetics, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
34
|
Ahmed HA, Ishrat T. The Brain AT2R-a Potential Target for Therapy in Alzheimer's Disease and Vascular Cognitive Impairment: a Comprehensive Review of Clinical and Experimental Therapeutics. Mol Neurobiol 2020; 57:3458-3484. [PMID: 32533467 PMCID: PMC8109287 DOI: 10.1007/s12035-020-01964-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/28/2020] [Indexed: 10/24/2022]
Abstract
Dementia is a potentially avertable tragedy, currently considered among the top 10 greatest global health challenges of the twenty-first century. Dementia not only robs individuals of their dignity and independence, it also has a ripple effect that starts with the inflicted individual's family and projects to the society as a whole. The constantly growing number of cases, along with the lack of effective treatments and socioeconomic impact, poses a serious threat to the sustainability of our health care system. Hence, there is a worldwide effort to identify new targets for the treatment of Alzheimer's disease (AD), the leading cause of dementia. Due to its multifactorial etiology and the recent clinical failure of several novel amyloid-β (Aβ) targeting therapies, a comprehensive "multitarget" approach may be most appropriate for managing this condition. Interestingly, renin angiotensin system (RAS) modulators were shown to positively impact all the factors involved in the pathophysiology of dementia including vascular dysfunction, Aβ accumulation, and associated cholinergic deficiency, in addition to tau hyperphosphorylation and insulin derangements. Furthermore, for many of these drugs, the preclinical evidence is also supported by epidemiological data and/or preliminary clinical trials. The purpose of this review is to provide a comprehensive update on the major causes of dementia including the risk factors, current diagnostic criteria, pathophysiology, and contemporary treatment strategies. Moreover, we highlight the angiotensin II receptor type 2 (AT2R) as an effective drug target and present ample evidence supporting its potential role and clinical applications in cognitive impairment to encourage further investigation in the clinical setting.
Collapse
Affiliation(s)
- Heba A Ahmed
- Department of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, 855 Monroe Avenue, Wittenborg Bldg, Room-231, Memphis, TN, 38163, USA
| | - Tauheed Ishrat
- Department of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, 855 Monroe Avenue, Wittenborg Bldg, Room-231, Memphis, TN, 38163, USA.
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA.
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
35
|
Khalifeh M, Read MI, Barreto GE, Sahebkar A. Trehalose against Alzheimer's Disease: Insights into a Potential Therapy. Bioessays 2020; 42:e1900195. [PMID: 32519387 DOI: 10.1002/bies.201900195] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 04/13/2020] [Indexed: 12/21/2022]
Abstract
Trehalose is a natural disaccharide with a remarkable ability to stabilize biomolecules. In recent years, trehalose has received growing attention as a neuroprotective molecule and has been tested in experimental models for different neurodegenerative diseases. Although the underlying neuroprotective mechanism of trehalose's action is unclear, one of the most important hypotheses is autophagy induction. The chaperone-like activity of trehalose and the ability to modulate inflammatory responses has also been reported. There is compelling evidence that the dysfunction of autophagy and aggregation of misfolded proteins contribute to the pathogenesis of Alzheimer's disease (AD) and other neurodegenerative disorders. Therefore, given the linking between trehalose and autophagy induction, it appears to be a promising therapy for AD. Herein, the published studies concerning the use of trehalose as a potential therapy for AD are summarized, providing a rationale for applying trehalose to reduce Alzheimer's pathology.
Collapse
Affiliation(s)
- Masoomeh Khalifeh
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Morgayn I Read
- Department of Pharmacology, School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
36
|
Huh TH, Yoon JL, Cho JJ, Kim MY, Ju YS. Survival Analysis of Patients with Alzheimer's Disease: A Study Based on Data from the Korean National Health Insurance Services' Senior Cohort Database. Korean J Fam Med 2020; 41:214-221. [PMID: 32321203 PMCID: PMC7385296 DOI: 10.4082/kjfm.18.0114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/10/2018] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Korea's rapidly aging population has experienced a sharp rise in the prevalence of dementia. Patients with Alzheimer's disease (AD), which is estimated to be about three-quarters of all patients with dementia, tend to have higher mortality rates compared with patients without Alzheimer's disease. In this study, a survival analysis of patients with AD was conducted in order to provide knowledge to those who provide medical care to these patients. METHODS Data on individuals over 65 years old in 2004 were extracted from the Korean National Health Insurance Services' Senior Cohort database (2002-2013). The subjects were 209,254 patients, including 2,695 who were first diagnosed with AD (the AD group) and 206,559 that had not been diagnosed with the disease (non-AD group). To investigate the independent effect of AD on survival, the Cox proportional-hazards model, hazard ratios (confidence interval of 95%), and the Kaplan-Meier method were used. RESULTS Mean survival time in the AD group was 5.3±3.3 years, which was about 2.5 years shorter than that in the non-AD group (7.8±2.4 years). The mortality rate in the AD group (66.3%) was higher than that in the non-AD group (26.3%). The adjusted hazard ratio in the AD group was 2.5 and, therefore, it was found that the AD group had a 2.5-fold higher risk of death than the non-AD group. CONCLUSION Overall, AD has a large, independent impact on survival. Survival time was shorter, and the mortality rate and risk were generally higher in the AD group, compared with the non-AD group.
Collapse
Affiliation(s)
- Tae Ho Huh
- Department of Family Medicine, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Korea
| | - Jong Lull Yoon
- Department of Family Medicine, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Korea.,Department of Family Medicine, Hallym University College of Medicine, Chuncheon, Korea
| | - Jung Jin Cho
- Department of Family Medicine, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Korea.,Department of Family Medicine, Hallym University College of Medicine, Chuncheon, Korea
| | - Mee Young Kim
- Department of Family Medicine, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Korea.,Department of Family Medicine, Hallym University College of Medicine, Chuncheon, Korea
| | - Young Soo Ju
- Department of Occupational and Environmental Medicine, Hallym University College of Medicine, Chuncheon, Korea
| |
Collapse
|
37
|
Dar KB, Bhat AH, Amin S, Reshi BA, Zargar MA, Masood A, Ganie SA. Elucidating Critical Proteinopathic Mechanisms and Potential Drug Targets in Neurodegeneration. Cell Mol Neurobiol 2020; 40:313-345. [PMID: 31584139 PMCID: PMC11449027 DOI: 10.1007/s10571-019-00741-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 08/06/2019] [Indexed: 12/18/2022]
Abstract
Neurodegeneration entails progressive loss of neuronal structure as well as function leading to cognitive failure, apathy, anxiety, irregular body movements, mood swing and ageing. Proteomic dysregulation is considered the key factor for neurodegeneration. Mechanisms involving deregulated processing of proteins such as amyloid beta (Aβ) oligomerization; tau hyperphosphorylation, prion misfolding; α-synuclein accumulation/lewy body formation, chaperone deregulation, acetylcholine depletion, adenosine 2A (A2A) receptor hyperactivation, secretase deregulation, leucine-rich repeat kinase 2 (LRRK2) mutation and mitochondrial proteinopathies have deeper implications in neurodegenerative disorders. Better understanding of such pathological mechanisms is pivotal for exploring crucial drug targets. Herein, we provide a comprehensive outlook about the diverse proteomic irregularities in Alzheimer's, Parkinson's and Creutzfeldt Jakob disease (CJD). We explicate the role of key neuroproteomic drug targets notably Aβ, tau, alpha synuclein, prions, secretases, acetylcholinesterase (AchE), LRRK2, molecular chaperones, A2A receptors, muscarinic acetylcholine receptors (mAchR), N-methyl-D-aspartate receptor (NMDAR), glial cell line-derived neurotrophic factor (GDNF) family ligands (GFLs) and mitochondrial/oxidative stress-related proteins for combating neurodegeneration and associated cognitive and motor impairment. Cross talk between amyloidopathy, synucleinopathy, tauopathy and several other proteinopathies pinpoints the need to develop safe therapeutics with ability to strike multiple targets in the aetiology of the neurodegenerative disorders. Therapeutics like microtubule stabilisers, chaperones, kinase inhibitors, anti-aggregation agents and antibodies could serve promising regimens for treating neurodegeneration. However, drugs should be target specific, safe and able to penetrate blood-brain barrier.
Collapse
Affiliation(s)
- Khalid Bashir Dar
- Department of Clinical Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
- Department of Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
| | - Aashiq Hussain Bhat
- Department of Clinical Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
- Department of Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
| | - Shajrul Amin
- Department of Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
| | - Bilal Ahmad Reshi
- Department of Biotechnology, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
| | - Mohammad Afzal Zargar
- Department of Clinical Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
| | - Akbar Masood
- Department of Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India
| | - Showkat Ahmad Ganie
- Department of Clinical Biochemistry, Faculty of Biological Sciences, University of Kashmir, Srinagar, India.
| |
Collapse
|
38
|
Husna Ibrahim N, Yahaya MF, Mohamed W, Teoh SL, Hui CK, Kumar J. Pharmacotherapy of Alzheimer's Disease: Seeking Clarity in a Time of Uncertainty. Front Pharmacol 2020; 11:261. [PMID: 32265696 PMCID: PMC7105678 DOI: 10.3389/fphar.2020.00261] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/24/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is recognized as a major health hazard that mostly affects people older than 60 years. AD is one of the biggest medical, economic, and social concerns to patients and their caregivers. AD was ranked as the 5th leading cause of global deaths in 2016 by the World Health Organization (WHO). Many drugs targeting the production, aggregation, and clearance of Aβ plaques failed to give any conclusive clinical outcomes. This mainly stems from the fact that AD is not a disease attributed to a single-gene mutation. Two hallmarks of AD, Aβ plaques and neurofibrillary tangles (NFTs), can simultaneously induce other AD etiologies where every pathway is a loop of consequential events. Therefore, the focus of recent AD research has shifted to exploring other etiologies, such as neuroinflammation and central hyperexcitability. Neuroinflammation results from the hyperactivation of microglia and astrocytes that release pro-inflammatory cytokines due to the neurological insults caused by Aβ plaques and NFTs, eventually leading to synaptic dysfunction and neuronal death. This review will report the failures and side effects of many anti-Aβ drugs. In addition, emerging treatments targeting neuroinflammation in AD, such as nonsteroidal anti-inflammatory drugs (NSAIDs) and receptor-interacting serine/threonine protein kinase 1 (RIPK1), that restore calcium dyshomeostasis and microglia physiological function in clearing Aβ plaques, respectively, will be deliberately discussed. Other novel pharmacotherapy strategies in treating AD, including disease-modifying agents (DMTs), repurposing of medications used to treat non-AD illnesses, and multi target-directed ligands (MTDLs) are also reviewed. These approaches open new doors to the development of AD therapy, especially combination therapy that can cater for several targets simultaneously, hence effectively slowing or stopping AD.
Collapse
Affiliation(s)
- Nurul Husna Ibrahim
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Mohamad Fairuz Yahaya
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Wael Mohamed
- Basic Medical Science Department, Kulliyyah of Medicine, International Islamic University Malaysia, Kuantan, Malaysia
- Faculty of Medicine, Department of Clinical Pharmacology, Menoufia University, Shebin El-Kom, Egypt
| | - Seong Lin Teoh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Chua Kien Hui
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
- Glycofood Sdn Bhd, Selangor, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| |
Collapse
|
39
|
Zhao Y, Long Z, Ding Y, Jiang T, Liu J, Li Y, Liu Y, Peng X, Wang K, Feng M, He G. Dihydroartemisinin Ameliorates Learning and Memory in Alzheimer's Disease Through Promoting Autophagosome-Lysosome Fusion and Autolysosomal Degradation for Aβ Clearance. Front Aging Neurosci 2020; 12:47. [PMID: 32210783 PMCID: PMC7067048 DOI: 10.3389/fnagi.2020.00047] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 02/11/2020] [Indexed: 01/07/2023] Open
Abstract
Dihydroartemisinin (DHA) is an active metabolite of sesquiterpene trioxane lactone extracted from Artemisia annua, which is used to treat malaria worldwide. DHA can activate autophagy, which is the main mechanism to remove the damaged cell components and recover the harmful or useless substances from eukaryotic cells and maintain cell viability through the autophagy lysosomal degradation system. Autophagy activation and autophagy flux correction are playing an important neuroprotective role in the central nervous system, as they accelerate the removal of toxic protein aggregates intracellularly and extracellularly to prevent neurodegenerative processes, such as Alzheimer's disease (AD). In this study, we explored whether this mechanism can mediate the neuroprotective effect of DHA on the AD model in vitro and in vivo. Three months of DHA treatment improved the memory and cognitive impairment, reduced the deposition of amyloid β plaque, reduced the levels of Aβ40 and Aβ42, and ameliorated excessive neuron apoptosis in APP/PS1 mice brain. In addition, DHA treatment increased the level of LC3 II/I and decreased the expression of p62. After Bafilomycin A1 and Chloroquine (CQ) blocked the fusion of autophagy and lysosome, as well as the degradation of autolysosomes (ALs), DHA treatment increased the level of LC3 II/I and decreased the expression of p62. These results suggest that DHA treatment can correct autophagic flux, improve autophagy dysfunction, inhibit abnormal death of neurons, promote the clearance of amyloid-β peptide (Aβ) fibrils, and have a multi-target effect on the neuropathological process, memory and cognitive deficits of AD.
Collapse
Affiliation(s)
- Yueyang Zhao
- Neuroscience Research Center, Chongqing Medical University, Chongqing, China
| | - Zhimin Long
- Neuroscience Research Center, Chongqing Medical University, Chongqing, China.,Department of Human Anatomy, Basic Medical School, Chongqing Medical University, Chongqing, China
| | - Ya Ding
- Neuroscience Research Center, Chongqing Medical University, Chongqing, China
| | - Tingting Jiang
- Neuroscience Research Center, Chongqing Medical University, Chongqing, China
| | - Jiajun Liu
- Neuroscience Research Center, Chongqing Medical University, Chongqing, China
| | - Yimin Li
- Neuroscience Research Center, Chongqing Medical University, Chongqing, China
| | - Yuanjie Liu
- Neuroscience Research Center, Chongqing Medical University, Chongqing, China.,Department of Human Anatomy, Basic Medical School, Chongqing Medical University, Chongqing, China
| | - Xuehua Peng
- Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Kejian Wang
- Neuroscience Research Center, Chongqing Medical University, Chongqing, China.,Department of Human Anatomy, Basic Medical School, Chongqing Medical University, Chongqing, China
| | - Min Feng
- Neuroscience Research Center, Chongqing Medical University, Chongqing, China
| | - Guiqiong He
- Neuroscience Research Center, Chongqing Medical University, Chongqing, China.,Department of Human Anatomy, Basic Medical School, Chongqing Medical University, Chongqing, China
| |
Collapse
|
40
|
Sahin L, Figueiro MG. Flickering Red-Light Stimulus for Promoting Coherent 40 Hz Neural Oscillation: A Feasibility Study. J Alzheimers Dis 2020; 75:911-921. [PMID: 32390635 PMCID: PMC8083946 DOI: 10.3233/jad-200179] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Coherent 40 Hz (gamma) neural oscillation indicates healthy brain activity and is known to be disrupted in Alzheimer's disease (AD) patients. 40 Hz entrainment by flickering light is known to significantly attenuate AD pathology in mice. OBJECTIVE To demonstrate the feasibility of using a lighting intervention to promote coherent 40 Hz neural oscillation, improved working memory performance, and reduced subjective sleepiness among a population of healthy young adults. If successful, the intervention could be extended to address cognitive impairment associated with mild cognitive impairment and AD. METHODS Nine healthy participants (median age 22 years, five females) were exposed to one of two lighting conditions per session in a within-subjects counterbalanced manner. The study's two sessions were separated by 1 week. Custom-built light masks provided either a 40 Hz flickering red light (FRL) intervention or a dark control condition (i.e., total darkness, light mask not energized) at participants' eyes. Data were collected four times per session: pre-exposure, after 25-min exposure, after 50-min exposure, and post-exposure. Each data collection period included a Karolinska Sleepiness Scale report, an electroencephalogram, and working memory (n-back) auditory performance testing. RESULTS The FRL intervention induced a significant increase in 40 Hz power and a modest increase in low gamma power. The intervention had no significant impact on working memory performance and subjective sleepiness compared to the control. However, increases in 40 Hz power were significantly correlated with reduced subjective sleepiness. CONCLUSION The results clearly demonstrate the feasibility of using a flickering light to increase 40 Hz power.
Collapse
Affiliation(s)
- Levent Sahin
- Lighting Research Center, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Mariana G Figueiro
- Lighting Research Center, Rensselaer Polytechnic Institute, Troy, NY, USA
| |
Collapse
|
41
|
De Simone A, Naldi M, Tedesco D, Bartolini M, Davani L, Andrisano V. Advanced analytical methodologies in Alzheimer’s disease drug discovery. J Pharm Biomed Anal 2020; 178:112899. [DOI: 10.1016/j.jpba.2019.112899] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 12/13/2022]
|
42
|
Kisby B, Jarrell JT, Agar ME, Cohen DS, Rosin ER, Cahill CM, Rogers JT, Huang X. Alzheimer's Disease and Its Potential Alternative Therapeutics. JOURNAL OF ALZHEIMER'S DISEASE & PARKINSONISM 2019; 9. [PMID: 31588368 PMCID: PMC6777730 DOI: 10.4172/2161-0460.1000477] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Alzheimer’s Disease (AD) is a chronic neurodegenerative disease that affects over 5 million individuals in the United States alone. Currently, there are only two kinds of pharmacological interventions available for symptomatic relief of AD; Acetyl Cholinesterase Inhibitors (AChEI) and N-methyl-D-aspartic Acid (NMDA) receptor antagonists and these drugs do not slow down or stop the progression of the disease. Several molecular targets have been implicated in the pathophysiology of AD, such as the tau (τ) protein, Amyloid-beta (Aβ), the Amyloid Precursor Protein (APP) and more and several responses have also been observed in the advancement of the disease, such as reduced neurogenesis, neuroinflammation, oxidative stress and iron overload. In this review, we discuss general features of AD and several small molecules across different experimental AD drug classes that have been studied for their effects in the context of the molecular targets and responses associated with the AD progression. These drugs include: Paroxetine, Desferrioxamine (DFO), N-acetylcysteine (NAC), Posiphen/-(−)Phenserine, JTR-009, Carvedilol, LY450139, Intravenous immunoglobulin G 10%, Indomethacin and Lithium Carbonate (Li2CO3).
Collapse
Affiliation(s)
- Brent Kisby
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Juliet T Jarrell
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - M Enes Agar
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - David S Cohen
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Eric R Rosin
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Catherine M Cahill
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Jack T Rogers
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Xudong Huang
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
43
|
Gutti G, Kakarla R, Kumar D, Beohar M, Ganeshpurkar A, Kumar A, Krishnamurthy S, Singh SK. Discovery of novel series of 2-substituted benzo[d]oxazol-5-amine derivatives as multi-target directed ligands for the treatment of Alzheimer's disease. Eur J Med Chem 2019; 182:111613. [PMID: 31437780 DOI: 10.1016/j.ejmech.2019.111613] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 01/04/2023]
Abstract
Alzheimer's disease (AD) is associated with multifactorial neuropathological conditions, which include cholinergic deficit, amyloid-beta plaques formation, loss of neuronal plasticity and neuronal death. Treating such multifactorial conditions with a single target directed approach is considered to be inadequate. Accordingly, multi-target directed ligand (MTDL) strategy has been evolved as an auspicious approach for the treatment of AD. In light of that, a library of 2-substituted benzo[d]oxazol-5-amine derivatives (29-39; 86-107) was designed using the scaffold hopping guided MTDLs strategy, synthesized and evaluated through various in-vitro and in-vivo biological studies. The optimal compound 92 exhibited potent inhibitory activities against AChE (IC50 = 0.052 ± 0.010 μM), BuChE (IC50 = 1.085 ± 0.035 μM), and significant amyloid-beta aggregation (20 μM) inhibition. The compound possessed better blood-brain barrier permeability (Pe = 10.80 ± 0.055 × 10-6 cm s-1) in PAMPA assay and neuro protective properties (40 μM) on SH-SY5Y neuroblastoma cell lines. Furthermore, in-vivo behavioural studies were performed on Y-maze test (scopolamine-induced amnesia model) and Morris water maze test (Aβ1-42 induced ICV rat model). The compound 92, at a dose of 10 mg/kg oral administration, demonstrated a substantial improvement of the cognitive and special memory impairment. In summary, both in-vitro and in-vivo investigations evidenced that compound 92 was a potential lead for the discovery of safe and effective disease-modifying agents for AD.
Collapse
Affiliation(s)
- Gopichand Gutti
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ramakrishna Kakarla
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Devendra Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Mahima Beohar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ankit Ganeshpurkar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ashok Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Sairam Krishnamurthy
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Sushil Kumar Singh
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India.
| |
Collapse
|
44
|
Duggal P, Mehan S. Neuroprotective Approach of Anti-Cancer Microtubule Stabilizers Against Tauopathy Associated Dementia: Current Status of Clinical and Preclinical Findings. J Alzheimers Dis Rep 2019; 3:179-218. [PMID: 31435618 PMCID: PMC6700530 DOI: 10.3233/adr-190125] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neuronal microtubule (MT) tau protein provides cytoskeleton to neuronal cells and plays a vital role including maintenance of cell shape, intracellular transport, and cell division. Tau hyperphosphorylation mediates MT destabilization resulting in axonopathy and neurotransmitter deficit, and ultimately causing Alzheimer’s disease (AD), a dementing disorder affecting vast geriatric populations worldwide, characterized by the existence of extracellular amyloid plaques and intracellular neurofibrillary tangles in a hyperphosphorylated state. Pre-clinically, streptozotocin stereotaxically mimics the behavioral and biochemical alterations similar to AD associated with tau pathology resulting in MT assembly defects, which proceed neuropathological cascades. Accessible interventions like cholinesterase inhibitors and NMDA antagonist clinically provides only symptomatic relief. Involvement of microtubule stabilizers (MTS) prevents tauopathy particularly by targeting MT oriented cytoskeleton and promotes polymerization of tubulin protein. Multiple in vitro and in vivo research studies have shown that MTS can hold substantial potential for the treatment of AD-related tauopathy dementias through restoration of tau function and axonal transport. Moreover, anti-cancer taxane derivatives and epothiolones may have potential to ameliorate MT destabilization and prevent the neuronal structural and functional alterations associated with tauopathies. Therefore, this current review strictly focuses on exploration of various clinical and pre-clinical features available for AD to understand the neuropathological mechanisms as well as introduce pharmacological interventions associated with MT stabilization. MTS from diverse natural sources continue to be of value in the treatment of cancer, suggesting that these agents have potential to be of interest in the treatment of AD-related tauopathy dementia in the future.
Collapse
Affiliation(s)
- Pallavi Duggal
- Neuropharmacology Division, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
45
|
Vico Varela E, Etter G, Williams S. Excitatory-inhibitory imbalance in Alzheimer's disease and therapeutic significance. Neurobiol Dis 2019; 127:605-615. [DOI: 10.1016/j.nbd.2019.04.010] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 04/08/2019] [Accepted: 04/12/2019] [Indexed: 11/29/2022] Open
|
46
|
Specific keratinase derived designer peptides potently inhibit Aβ aggregation resulting in reduced neuronal toxicity and apoptosis. Biochem J 2019; 476:1817-1841. [DOI: 10.1042/bcj20190183] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/22/2019] [Accepted: 05/27/2019] [Indexed: 11/17/2022]
Abstract
Abstract
Compelling evidence implicates self-assembly of amyloid-β (Aβ1–42) peptides into soluble oligomers and fibrils as a major underlying event in Alzheimer's disease (AD) pathogenesis. Herein, we employed amyloid-degrading keratinase (kerA) enzyme as a key Aβ1–42-binding scaffold to identify five keratinase-guided peptides (KgPs) capable of interacting with and altering amyloidogenic conversion of Aβ1–42. The KgPs showed micromolar affinities with Aβ1–42 and abolished its sigmoidal amyloidogenic transition, resulting in abrogation of fibrillogenesis. Comprehensive assessment using dynamic light scattering (DLS), atomic force microscopy (AFM) and Fourier-transform infrared (FTIR) spectroscopy showed that KgPs induced the formation of off-pathway oligomers comparatively larger than the native Aβ1–42 oligomers but with a significantly reduced cross-β signature. These off-pathway oligomers exhibited low immunoreactivity against oligomer-specific (A11) and fibril-specific (OC) antibodies and rescued neuronal cells from Aβ1–42 oligomer toxicity as well as neuronal apoptosis. Structural analysis using molecular docking and molecular dynamics (MD) simulations showed two preferred KgP binding sites (Lys16–Phe20 and Leu28–Val39) on the NMR ensembles of monomeric and fibrillar Aβ1–42, indicating an interruption of crucial hydrophobic and aromatic interactions. Overall, our results demonstrate a new approach for designing potential anti-amyloid molecules that could pave way for developing effective therapeutics against AD and other amyloid diseases.
Collapse
|
47
|
Li Z, Yang N, Lei X, Lin C, Li N, Jiang X, Wei X, Xu B. The association between the ApoE polymorphisms and the MRI-defined intracranial lesions in a cohort of southern China population. J Clin Lab Anal 2019; 33:e22950. [PMID: 31199015 PMCID: PMC6757122 DOI: 10.1002/jcla.22950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 05/13/2019] [Accepted: 05/23/2019] [Indexed: 11/20/2022] Open
Abstract
Background The apolipoprotein E (APOE) ε4 allele is considered as a risk factor for Alzheimer's disease (AD). However, the association of APOE allele with MRI evidence of intracranial lesions has not been well understood. Methods Quantitative real‐time PCR was performed to detect the APOE genotype; MRI was examined for intracranial lesions. Their association was evaluated in a cohort of 226 AD patients and 2607 healthy individuals in southern China. Results The frequencies of ε2, ε3, and ε4 alleles were 8.0%, 82.9%, and 9.1% in the whole study population. The frequency of APOE‐ε4 allele was significantly higher in the AD subjects than that in the control group (14.4% vs 8.6%, P < 0.001). We found that brain atrophy occurred at a rate of 12.3% in ε4 allele group vs 8.5% in non‐ε4 genotype group, with a significance of P = 0.008. Severe brain atrophy occurred at a rate of 1.0% in ε4 allele group vs 0.2% in non‐ε4 genotype group (P = 0.011). The individuals carrying APOE ε4/ε4 had an odds ratio (OR) of 7.64 (P < 0.01) for developing AD, while the APOE ε3/ε4 gene carriers had an OR of 1.47 (P = 0.031) and the OR in APOE ε2/ε3 carriers is 0.81 (P = 0.372). Interestingly, we found that the risk of ε4/ε4 allele carrier developing AD was significantly higher in male (P < 0.001) than female (P = 0.478). Conclusion Compared to ε2 and ε3 alleles, the presence of APOE‐ε4 allele might increase the risk for AD in a dose‐dependent manner in southern China. Moreover, the presence of APOE‐ε4 allele results in a higher incidence of brain atrophy.
Collapse
Affiliation(s)
- Zhuoran Li
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Na Yang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiuxia Lei
- Department of Laboratory Medicine, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chuying Lin
- Department of Laboratory Medicine, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Nan Li
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinqing Jiang
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xinhua Wei
- Department of Radiology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Banglao Xu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
48
|
Gómez-Gómez ME, Zapico SC. Frailty, Cognitive Decline, Neurodegenerative Diseases and Nutrition Interventions. Int J Mol Sci 2019; 20:ijms20112842. [PMID: 31212645 PMCID: PMC6600148 DOI: 10.3390/ijms20112842] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 06/01/2019] [Accepted: 06/05/2019] [Indexed: 12/12/2022] Open
Abstract
Currently the human population is aging faster. This leads to higher dependency rates and the transformation of health and social care to adapt to this aged population. Among the changes developed by this population is frailty. It is defined as a clinically detectable syndrome, related to the aging of multiple physiological systems, which prompts a situation of vulnerability. The etiology of frailty seems to be multifactorial and its pathophysiology is influenced by the interaction of numerous factors. Morley et al. propose four main mechanisms triggering the frailty: atherosclerosis, sarcopenia, cognitive deterioration and malnutrition, with their respective metabolic alterations. Malnutrition is associated with cognitive impairment or functional loss, but it is also known that an inadequate nutritional status predisposes to cognitive frailty. Additionally, nutritional factors that may influence vascular risk factors will potentially have an effect on dementia decline among patients with cognitive frailty. This review aims to describe the nutritional factors that have been researched so far which may lead to the development of frailty, and especially cognitive decline.
Collapse
Affiliation(s)
| | - Sara C Zapico
- International Forensic Research Institute and Chemistry Department, Florida International University, 11200 SW 8 St., CP323, Miami, FL 33199, USA.
- Anthropology Department, Smithsonian Institution, NMNH, MRC 112, 10th and Constitution Ave, NW, PO Box 37012, Washington, DC 20560, USA.
| |
Collapse
|
49
|
Abstract
AbstractIn recent decades, clinical trials in Alzheimer’s disease (AD) have failed at an unprecedented rate. The etiology of AD has since come under renewed scrutiny, both to elucidate the underlying pathologies and to identify novel therapeutic strategies. Here, diet has emerged as a potential causative/protective agent. A variety of nutrients, including lipids, minerals, vitamins, antioxidants and sugars as well as broader dietary patterns and microbiotal interactions have demonstrated associations with AD. Although clinical trials have yet to definitively implicate any singular dietary element as therapeutic or causative, it is apparent that dietary preferences, likely in complex synergies, may influence the risk, onset and course of AD. This review catalogs the impact of major dietary elements on AD. It further examines an unexplored reciprocal association where AD may modulate diet, as well as how potential therapeutics may complicate these interactions. In doing so, we observe diet may have profound effects on the outcome of a clinical trial, either as a confounder of a drug/disease interaction or as a generally disruptive covariate. We therefore conclude that future clinical trials in AD should endeavor to control for diet, either in study design or subsequent analyses.
Collapse
|
50
|
Vidal C, Daescu K, Fitzgerald KE, Starokadomska A, Bezprozvanny I, Zhang L. Amyloid β perturbs elevated heme flux induced with neuronal development. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2019; 5:27-37. [PMID: 30723777 PMCID: PMC6352316 DOI: 10.1016/j.trci.2018.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Introduction Heme is a central molecule in mitochondrial respiration and ATP generation in neuronal cells. Thus, we assessed the importance of altered heme metabolism in Alzheimer's disease (AD) pathogenesis. Methods To investigate the role of altered heme metabolism in AD, we identified heme-related proteins whose expression is altered in AD patients and mouse models exhibiting amyloid pathology. We detected the levels of proteins involved in heme synthesis, uptake, degradation, and function during neuronal differentiation and characterized the effects of Aβ. Results We found that the expression levels of the rate-limiting heme synthetic enzyme ALAS1 and heme degradation enzyme HO-2 are selectively decreased in AD patients and mice. Aβ selectively reduces the levels of HO-2 and heme degradation, which are elevated to support neuronal functions in fully differentiated neuronal cells. Discussion Our data show that lowered heme metabolism, particularly the decreased levels of heme degradation and HO-2, is likely a very early event in AD pathogenesis.
Collapse
Affiliation(s)
- Chantal Vidal
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Kelly Daescu
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Keely E Fitzgerald
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Anna Starokadomska
- Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ilya Bezprozvanny
- Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Li Zhang
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|