1
|
Kelliny S, Zhou X, Bobrovskaya L. Alzheimer's Disease and Frontotemporal Dementia: A Review of Pathophysiology and Therapeutic Approaches. J Neurosci Res 2025; 103:e70046. [PMID: 40387258 PMCID: PMC12087441 DOI: 10.1002/jnr.70046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/01/2025] [Accepted: 05/02/2025] [Indexed: 05/20/2025]
Abstract
Alzheimer's disease (AD) is a devastating form of dementia, with the number of affected individuals rising sharply. The main hallmarks of the disease include amyloid-beta plaque deposits and neurofibrillary tangles consisting of hyperphosphorylated tau protein, besides other pathological features that contribute to the disease's complexity. The causes of sporadic AD are multifactorial and mostly age-related and involve risk factors such as diabetes and cardiovascular or cerebrovascular disorders. Frontotemporal dementia (FTD) is another type of dementia characterized by a spectrum of behaviors, memory, and motor abnormalities and associated with abnormal depositions of protein aggregation, including tau protein. Currently approved medications are symptomatic, and no disease-modifying therapy is available to halt the disease progression. Therefore, the development of multi-targeted therapeutic approaches could hold promise for the treatment of AD and other neurodegenerative disorders, including tauopathies. In this article, we will discuss the pathophysiology of AD and FTD, the proposed hypotheses, and current therapeutic approaches, highlighting the development of novel drug candidates and the progress of clinical trials in this field of research.
Collapse
Affiliation(s)
- Sally Kelliny
- Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
- Faculty of PharmacyAssiut UniversityAssiutEgypt
| | - Xin‐Fu Zhou
- Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Larisa Bobrovskaya
- Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| |
Collapse
|
2
|
Devarakonda SS, Basha S, Pithakumar A, L B T, Mukunda DC, Rodrigues J, K A, Biswas S, Pai AR, Belurkar S, Mahato KK. Molecular mechanisms of neurofilament alterations and its application in assessing neurodegenerative disorders. Ageing Res Rev 2024; 102:102566. [PMID: 39481763 DOI: 10.1016/j.arr.2024.102566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/04/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024]
Abstract
Neurofilaments are intermediate filaments present in neurons. These provide structural support and maintain the size and shape of the neurons. Dysregulation, mutation, and aggregation of neurofilaments raise the levels of these proteins in the blood and cerebrospinal fluid (CSF), which are characteristic features of axonal damage and certain rare neurological diseases, such as Giant Axonal Neuropathy and Charcot-Mare-Tooth disease. Understanding the structure, dynamics, and function of neurofilaments has been greatly enhanced by a diverse range of biochemical and preclinical investigations conducted over more than four decades. Recently, there has been a resurgence of interest in post-translational modifications of neurofilaments, such as phosphorylation, aggregation, mutation, oxidation, etc. Over the past twenty years, several rare disorders have been studied from structural alterations of neurofilaments. These disorders are monitored by fluid biomarkers such as neurofilament light chains. Currently, there are many tools, such as Enzyme-Linked Immunosorbent Assay, Electrochemiluminescence Assay, Single-Molecule Array, Western/immunoblotting, etc., in use to assess the neurofilament proteins in Blood and CSF. However, all these techniques utilize expensive, non-specific, or antibody-based methods, which make them unsuitable for routine screening of neurodegenerative disorders. This provides room to search for newer sensitive, cost-effective, point-of-care tools for rapid screening of the disease. For a long time, the molecular mechanisms of neurofilaments have been poorly understood due to insufficient research attempts, and a deeper understanding of them remains elusive. Therefore, this review aims to highlight the available literature on molecular mechanisms of neurofilaments and the function of neurofilaments in axonal transport, axonal conduction, axonal growth, and neurofilament aggregation, respectively. Further, this review discusses the role of neurofilaments as potential biomarkers for the identification of several neurodegenerative diseases in clinical laboratory practice.
Collapse
Affiliation(s)
| | - Shaik Basha
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Anjana Pithakumar
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Thoshna L B
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | | | - Jackson Rodrigues
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Ameera K
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Shimul Biswas
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Aparna Ramakrishna Pai
- Department of Neurology, Kasturba Medical College-Manipal, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Sushma Belurkar
- Department of Pathology, Kasturba Medical College-Manipal, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India
| | - Krishna Kishore Mahato
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal - 576104, Karnataka, India.
| |
Collapse
|
3
|
Song QH, Zhao KX, Huang S, Chen T, He L. Escape from X-chromosome inactivation and sex differences in Alzheimer's disease. Rev Neurosci 2024; 35:341-354. [PMID: 38157427 DOI: 10.1515/revneuro-2023-0108] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/24/2023] [Indexed: 01/03/2024]
Abstract
Sex differences exist in the onset and progression of Alzheimer's disease. Globally, women have a higher prevalence, while men with Alzheimer's disease experience earlier mortality and more pronounced cognitive decline than women. The cause of sex differences in Alzheimer's disease remains unclear. Accumulating evidence suggests the potential role of X-linked genetic factors in the sex difference of Alzheimer's disease (AD). During embryogenesis, a remarkable process known as X-chromosome inactivation (XCI) occurs in females, leading to one of the X chromosomes undergoing transcriptional inactivation, which balances the effects of two X chromosomes in females. Nevertheless, certain genes exceptionally escape from XCI, which provides a basis for dual expression dosage of specific genes in females. Based on recent research findings, we explore key escape genes and their potential therapeutic use associated with Alzheimer's disease. Also, we discuss their possible role in driving the sex differences in Alzheimer's disease. This will provide new perspectives for precision medicine and gender-specific treatment of AD.
Collapse
Affiliation(s)
- Qing-Hua Song
- Department of Pharmacology, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing 210009, Jiangsu Province, China
| | - Ke-Xuan Zhao
- Department of Pharmacology, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing 210009, Jiangsu Province, China
| | - Shuai Huang
- Department of Pharmacology, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing 210009, Jiangsu Province, China
| | - Tong Chen
- Department of Pharmacology, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing 210009, Jiangsu Province, China
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing 210009, Jiangsu Province, China
| |
Collapse
|
4
|
Iranpanah A, Fakhri S, Bahrami G, Majnooni MB, Gravandi MM, Taghavi S, Badrbani MA, Amirian R, Farzaei MH. Protective effect of a hydromethanolic extract from Fraxinus excelsior L. bark against a rat model of aluminum chloride-induced Alzheimer's disease: Relevance to its anti-inflammatory and antioxidant effects. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117708. [PMID: 38181932 DOI: 10.1016/j.jep.2024.117708] [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: 10/25/2023] [Revised: 12/23/2023] [Accepted: 01/02/2024] [Indexed: 01/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fraxinus excelsior L. (FE), commonly known as the ash, belongs to the Oleaceae family and has shown several pharmacological and biological properties, such as antioxidant, immunomodulatory, neuroprotective, and anti-inflammatory effects. It has also attracted the most attention toward neuroinflammation. Moreover, FE bark and leaves have been used to treat neurological disorders, aging, neuropathic pain, urinary complaints, and articular pain in traditional and ethnomedicine. Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder resulting from the involvement of amyloid-beta, metal-induced oxidative stress, and neuroinflammation. AIM OF THE STUDY The objective of the current study was to assess the neuroprotective effects of hydromethanolic extract from FE bark in an AlCl3-induced rat model of AD. MATERIALS AND METHODS The maceration process was utilized to prepare the hydromethanolic extract of FE bark, and characterized by LC-MS/MS. To assess the anti-AD effects of the FE extract, rats were categorized into five different groups, AlCl3; normal control; FE-treated groups at 50, 100, and 200 mg/kg. Passive avoidance learning test, Y-maze, open field, and elevated plus maze behavioral tests were evaluated on days 7 and 14 to analyze the cognitive impairments. Zymography analysis, biochemical tests, and histopathological changes were also followed in different groups. RESULTS LC-MS/MS analysis indicated the presence of coumarins, including isofraxidin7-O-diglucoside in the methanolic extract of FE as a new isofraxidin derivative in this genus. FE significantly improved memory and cognitive function, maintained weight, prevented neuronal damages, and preserved the hippocampus's histological features, as demonstrated by behavioral tests and histopathological analysis. FE increased anti-inflammatory MMP-2 activity, whereas it decreased that of inflammatory MMP-9. Moreover, FE increased plasma antioxidant capacity by enhancing CAT and GSH while decreasing nitrite levels in the serum of treated groups. In comparison between the treated groups, the rats that received high doses of the FE extract (200 mg/kg) showed the highest therapeutic effect. CONCLUSION FE rich in coumarins could be an effective anti-AD adjunct agent, passing through antioxidant and anti-inflammatory pathways. These results encourage further studies for the development of this extract as a promising agent in preventing, managing, or treating AD and related diseases.
Collapse
Affiliation(s)
- Amin Iranpanah
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Gholamreza Bahrami
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Bagher Majnooni
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Sara Taghavi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Mehdi Azadi Badrbani
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Roshanak Amirian
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
5
|
Pena E, San Martin-Salamanca R, El Alam S, Flores K, Arriaza K. Tau Protein Alterations Induced by Hypobaric Hypoxia Exposure. Int J Mol Sci 2024; 25:889. [PMID: 38255962 PMCID: PMC10815386 DOI: 10.3390/ijms25020889] [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: 11/15/2023] [Revised: 12/21/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Tauopathies are a group of neurodegenerative diseases whose central feature is dysfunction of the microtubule-associated protein tau (MAPT). Although the exact etiology of tauopathies is still unknown, it has been hypothesized that their onset may occur up to twenty years before the clear emergence of symptoms, which has led to questions about whether the prognosis of these diseases can be improved by, for instance, targeting the factors that influence tauopathy development. One such factor is hypoxia, which is strongly linked to Alzheimer's disease because of its association with obstructive sleep apnea and has been reported to affect molecular pathways related to the dysfunction and aggregation of tau proteins and other biomarkers of neurological damage. In particular, hypobaric hypoxia exposure increases the activation of several kinases related to the hyperphosphorylation of tau in neuronal cells, such as ERK, GSK3β, and CDK5. In addition, hypoxia also increases the levels of inflammatory molecules (IL-β1, IL-6, and TNF-α), which are also associated with neurodegeneration. This review discusses the many remaining questions regarding the influence of hypoxia on tauopathies and the contribution of high-altitude exposure to the development of these diseases.
Collapse
Affiliation(s)
| | | | - Samia El Alam
- High Altitude Medicine Research Center (CEIMA), Arturo Prat University, Iquique 1110939, Chile; (E.P.); (R.S.M.-S.); (K.F.); (K.A.)
| | | | | |
Collapse
|
6
|
Qiu F, Wang Y, Du Y, Zeng C, Liu Y, Pan H, Ke C. Current evidence for J147 as a potential therapeutic agent in nervous system disease: a narrative review. BMC Neurol 2023; 23:317. [PMID: 37674139 PMCID: PMC10481599 DOI: 10.1186/s12883-023-03358-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 08/03/2023] [Indexed: 09/08/2023] Open
Abstract
Curcumin has anti-inflammatory, antioxidant, and anticancer effects and is used to treat diseases such as dermatological diseases, infection, stress, depression, and anxiety. J147, an analogue of curcumin, is designed and synthesized with better stability and bioavailability. Accumulating evidence demonstrates the potential role of J147 in the prevention and treatment of Alzheimer's disease, diabetic neuropathy, ischemic stroke, depression, anxiety, and fatty liver disease. In this narrative review, we summarized the background and biochemical properties of J147 and discussed the role and mechanism of J147 in different diseases. Overall, the mechanical attributes of J147 connote it as a potential target for the prevention and treatment of neurological diseases.
Collapse
Affiliation(s)
- Fang Qiu
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, Guangdong, China
- Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China
| | - Yanmei Wang
- Department of critical care medicine, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong, China
| | - Yunbo Du
- Department of critical care medicine, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong, China
| | - Changchun Zeng
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, Guangdong, China
| | - Yuqiang Liu
- Department of Anesthesiology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518025, Guangdong, China.
| | - Haobo Pan
- Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China.
| | - Changneng Ke
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, Guangdong, China.
| |
Collapse
|
7
|
Valverde-Salazar V, Ruiz-Gabarre D, García-Escudero V. Alzheimer's Disease and Green Tea: Epigallocatechin-3-Gallate as a Modulator of Inflammation and Oxidative Stress. Antioxidants (Basel) 2023; 12:1460. [PMID: 37507998 PMCID: PMC10376369 DOI: 10.3390/antiox12071460] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/05/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, characterised by a marked decline of both memory and cognition, along with pathophysiological hallmarks including amyloid beta peptide (Aβ) accumulation, tau protein hyperphosphorylation, neuronal loss and inflammation in the brain. Additionally, oxidative stress caused by an imbalance between free radicals and antioxidants is considered one of the main risk factors for AD, since it can result in protein, lipid and nucleic acid damage and exacerbate Aβ and tau pathology. To date, there is a lack of successful pharmacological approaches to cure or even ameliorate the terrible impact of this disease. Due to this, dietary compounds with antioxidative and anti-inflammatory properties acquire special relevance as potential therapeutic agents. In this context, green tea, and its main bioactive compound, epigallocatechin-3-gallate (EGCG), have been targeted as a plausible option for the modulation of AD. Specifically, EGCG acts as an antioxidant by regulating inflammatory processes involved in neurodegeneration such as ferroptosis and microglia-induced cytotoxicity and by inducing signalling pathways related to neuronal survival. Furthermore, it reduces tau hyperphosphorylation and aggregation and promotes the non-amyloidogenic route of APP processing, thus preventing the formation of Aβ and its subsequent accumulation. Taken together, these results suggest that EGCG may be a suitable candidate in the search for potential therapeutic compounds for neurodegenerative disorders involving inflammation and oxidative stress, including Alzheimer's disease.
Collapse
Affiliation(s)
- Víctor Valverde-Salazar
- Department of Anatomy, Histology and Neuroscience, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | - Daniel Ruiz-Gabarre
- Department of Anatomy, Histology and Neuroscience, School of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | - Vega García-Escudero
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, 28031 Madrid, Spain
- Institute for Molecular Biology-IUBM, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
8
|
Choi Y, Shin S, Son HJ, Lee NH, Myeong SH, Lee C, Jang H, Choi SJ, Kim HJ, Na DL. Identification of potential biomarkers related to mesenchymal stem cell response in patients with Alzheimer's disease. Stem Cell Res Ther 2023; 14:178. [PMID: 37468918 PMCID: PMC10357744 DOI: 10.1186/s13287-023-03410-8] [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: 07/01/2022] [Accepted: 07/06/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND Preclinical studies showed that mesenchymal stem cells (MSCs) ameliorate tau phosphorylation, amyloid-beta accumulation, and inflammation in Alzheimer's disease (AD) mouse models via secretion of neurotrophic factors and cytokines. We aimed to identify CSF biomarkers that can be used to predict or monitor the response to MSCs in patients with AD. METHODS AD patients were injected with human umbilical cord blood-MSCs (n = 22) or placebo (n = 12). The cerebrospinal fluid (CSF) samples were collected at baseline, one day after the first injection, and one day after the third injection. The patients injected with MSCs were classified into good responder (GR) or poor responder (PR) groups based on the rate of changes in the ratio of total-tau and phosphorylated-tau in the CSF. We selected three typical participants in each group, and their CSF protein levels were analyzed using liquid chromatography/tandem mass spectrometry (LC-MS/MS). RESULTS In the LC-MS/MS analysis, 1,667 proteins were identified. Eleven proteins showed significant differences between the typical GR and PR at baseline. Based on their significance level and known functions, two proteins, reticulocalbin-3 (RCN3) and follistatin-related protein 3 (FSTL3), were selected as potential biomarkers to predict MSC response. A total of 173 proteins showed significant change one day after the third injection compared to the baseline in typical GR. We excluded 45 proteins that showed significant change after the third injection compared to the baseline in the typical PR. Based on their significance level and known function, four proteins, scrapie-responsive protein 1 (SCRG1), neural proliferation differentiation and control protein (NPDC1), apolipoprotein E (ApoE), and cystatin C (CysC), were selected as potential biomarker to monitor MSC response. Additionally, functional analysis revealed that the increased CSF proteins after the third injection compared to the baseline in the typical GR were associated with synaptogenesis. CONCLUSIONS This study identified two proteins (RCN3 and FSTL3) that may be potential biomarkers for predicting MSC response and four proteins (SCRG1, NPDC1, ApoE, CysC) that may be potential biomarkers for monitoring MSC response in patients with AD. Further studies are needed to validate our results. Trial registration Clinical Trials.gov, NCT02054208. Registered on 4 February 2014. Samsung Medical Center IRB File No.2017-04-025. Registered on 20 June 2017.
Collapse
Affiliation(s)
- Yejoo Choi
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Sungho Shin
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Hyo Jin Son
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- School of Medicine, Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Neuroscience Center, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Na-Hee Lee
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06355, Republic of Korea
| | - Su Hyeon Myeong
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Cheolju Lee
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Hyemin Jang
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Neuroscience Center, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Soo Jin Choi
- Biomedical Research Institute, MEDIPOST Co., Ltd., 21, Daewangpangyo-ro 644 Beon-gil, Seongnam-si, Gyeonggi-do, 13494, Republic of Korea
| | - Hee Jin Kim
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea.
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
- School of Medicine, Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
- Neuroscience Center, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06355, Republic of Korea.
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea.
| | - Duk L Na
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea.
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Xiang L, Wang Y, Liu S, Liu B, Jin X, Cao X. Targeting Protein Aggregates with Natural Products: An Optional Strategy for Neurodegenerative Diseases. Int J Mol Sci 2023; 24:11275. [PMID: 37511037 PMCID: PMC10379780 DOI: 10.3390/ijms241411275] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Protein aggregation is one of the hallmarks of aging and aging-related diseases, especially for the neurodegenerative diseases (NDs) such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), Amyotrophic lateral sclerosis (ALS), and others. In these diseases, many pathogenic proteins, such as amyloid-β, tau, α-Syn, Htt, and FUS, form aggregates that disrupt the normal physiological function of cells and lead to associated neuronal lesions. Protein aggregates in NDs are widely recognized as one of the important targets for the treatment of these diseases. Natural products, with their diverse biological activities and rich medical history, represent a great treasure trove for the development of therapeutic strategies to combat disease. A number of in vitro and in vivo studies have shown that natural products, by virtue of their complex molecular scaffolds that specifically bind to pathogenic proteins and their aggregates, can inhibit the formation of aggregates, disrupt the structure of aggregates and destabilize them, thereby alleviating conditions associated with NDs. Here, we systematically reviewed studies using natural products to improve disease-related symptoms by reducing or inhibiting the formation of five pathogenic protein aggregates associated with NDs. This information should provide valuable insights into new directions and ideas for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Lingzhi Xiang
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou 311300, China
| | - Yanan Wang
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou 311300, China
| | - Shenkui Liu
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou 311300, China
| | - Beidong Liu
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou 311300, China
- Department of Chemistry and Molecular Biology, University of Gothenburg, 41390 Gothenburg, Sweden
| | - Xuejiao Jin
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou 311300, China
| | - Xiuling Cao
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Hangzhou 311300, China
| |
Collapse
|
10
|
Tavakoli M, Ghadami SA, Adibi H, Gulcan HO. Synthesis of benzylidene-benzofuranone derivatives as probes for detection of amyloid fibrils in cells. J Biomol Struct Dyn 2023; 41:14989-15002. [PMID: 36866639 DOI: 10.1080/07391102.2023.2184635] [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: 10/28/2022] [Accepted: 02/20/2023] [Indexed: 03/04/2023]
Abstract
Aggregated protein is the common cause of a wide variety of neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease, etc. It is proven that protein aggregation like amyloid β (Aβ) is one of the critical factors causing AD and, its diagnosis in the early stages of the disease is important for the treatment or prevention of AD. To have a better understanding of protein aggregation and its pathologies, there is a huge need to design and develop new and more trustworthy probe molecules for in vitro amyloid quantification and in vivo amyloid imaging. In this study, 17 new biomarker compounds, have been synthesized from benzofuranone derivatives, to detect and identify amyloid in vitro (dye-binding assay) as well as in the cell by staining method. According to the results, some of these synthetic derivatives can be considered suitable identifiers and quantifiers to detect amyloid fibrils in vitro. Compared to thioflavin T, 4 probes out of 17 probes have shown good results in selectivity and detectability of Aβ depositions, and their binding properties were also confirmed with in silico analysis. The drug-likeness prediction results for selected compounds by the Swiss ADME server show a satisfactory percentage of blood-brain barrier (BBB) permeability and gastrointestinal (GI) absorption. Among all of them, compound 10 was able to show better binding properties than others, and in vivo study showed that this compound was capable of detecting intracellular amyloid.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mohsen Tavakoli
- Department of Biotechnology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Faculty of Pharmacy, Eastern Mediterranean University, TRNC, Famagusta, Turkey
| | | | - Hadi Adibi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | |
Collapse
|
11
|
Moore KBE, Hung TJ, Fortin JS. Hyperphosphorylated tau (p-tau) and drug discovery in the context of Alzheimer's disease and related tauopathies. Drug Discov Today 2023; 28:103487. [PMID: 36634842 PMCID: PMC9975055 DOI: 10.1016/j.drudis.2023.103487] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 12/14/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, characterized by intracellular neurofibrillary tangles (NFTs) and extracellular β-amyloid (βA) plaques. No disease-modifying therapy is currently available to prevent the progression of, or cure, the disease. Misfolded hyperphosphorylated tau (p-tau) is considered a pivotal point in the pathogenesis of AD and other tauopathies. Compelling evidence suggests that it is a key driver of the accumulation of NFTs and can be directly correlated with the extent of dementia in patients with AD. Therefore, inhibiting tau hyperphosphorylation-induced aggregation could be a viable strategy to discover and develop therapeutics for patients with AD.
Collapse
Affiliation(s)
- Kendall B E Moore
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN, USA
| | - Ta-Jung Hung
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN, USA
| | - Jessica S Fortin
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, 625 Harrison Street, West Lafayette, IN, USA.
| |
Collapse
|
12
|
Frigerio I, Laansma MA, Lin CP, Hermans EJM, Bouwman MMA, Bol JGJM, Galis-de Graaf Y, Hepp DH, Rozemuller AJM, Barkhof F, van de Berg WDJ, Jonkman LE. Neurofilament light chain is increased in the parahippocampal cortex and associates with pathological hallmarks in Parkinson's disease dementia. Transl Neurodegener 2023; 12:3. [PMID: 36658627 PMCID: PMC9854202 DOI: 10.1186/s40035-022-00328-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/17/2022] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Increased neurofilament levels in biofluids are commonly used as a proxy for neurodegeneration in several neurodegenerative disorders. In this study, we aimed to investigate the distribution of neurofilaments in the cerebral cortex of Parkinson's disease (PD), PD with dementia (PDD) and dementia with Lewy bodies (DLB) donors, and its association with pathology load and MRI measures of atrophy and diffusivity. METHODS Using a within-subject post-mortem MRI-pathology approach, we included 9 PD, 12 PDD/DLB and 18 age-matched control donors. Cortical thickness and mean diffusivity (MD) metrics were extracted respectively from 3DT1 and DTI at 3T in-situ MRI. After autopsy, pathological hallmarks (pSer129-αSyn, p-tau and amyloid-β load) together with neurofilament light-chain (NfL) and phosphorylated-neurofilament medium- and heavy-chain (p-NfM/H) immunoreactivity were quantified in seven cortical regions, and studied in detail with confocal-laser scanning microscopy. The correlations between MRI and pathological measures were studied using linear mixed models. RESULTS Compared to controls, p-NfM/H immunoreactivity was increased in all cortical regions in PD and PDD/DLB, whereas NfL immunoreactivity was increased in the parahippocampal and entorhinal cortex in PDD/DLB. NfL-positive neurons showed degenerative morphological features and axonal fragmentation. The increased p-NfM/H correlated with p-tau load, and NfL correlated with pSer129-αSyn but more strongly with p-tau load in PDD/DLB. Lastly, neurofilament immunoreactivity correlated with cortical thinning in PD and with increased cortical MD in PDD/DLB. CONCLUSIONS Taken together, increased neurofilament immunoreactivity suggests underlying axonal injury and neurofilament accumulation in morphologically altered neurons with increased pathological burden. Importantly, we demonstrate that such neurofilament markers at least partly explain MRI measures that are associated with the neurodegenerative process.
Collapse
Affiliation(s)
- Irene Frigerio
- Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1118, Amsterdam, The Netherlands. .,Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands. .,Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands.
| | - Max A. Laansma
- grid.12380.380000 0004 1754 9227Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1118, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| | - Chen-Pei Lin
- grid.12380.380000 0004 1754 9227Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1118, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| | - Emma J. M. Hermans
- grid.12380.380000 0004 1754 9227Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1118, Amsterdam, The Netherlands
| | - Maud M. A. Bouwman
- grid.12380.380000 0004 1754 9227Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1118, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| | - John G. J. M. Bol
- grid.12380.380000 0004 1754 9227Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1118, Amsterdam, The Netherlands
| | - Yvon Galis-de Graaf
- grid.12380.380000 0004 1754 9227Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1118, Amsterdam, The Netherlands
| | - Dagmar H. Hepp
- grid.12380.380000 0004 1754 9227Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1118, Amsterdam, The Netherlands ,grid.12380.380000 0004 1754 9227Department of Neurology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Annemieke J. M. Rozemuller
- grid.12380.380000 0004 1754 9227Department of Pathology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Frederik Barkhof
- grid.484519.5Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands ,grid.12380.380000 0004 1754 9227Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands ,grid.83440.3b0000000121901201Institutes of Neurology and Healthcare Engineering, University College London, London, UK
| | - Wilma D. J. van de Berg
- grid.12380.380000 0004 1754 9227Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1118, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Laura E. Jonkman
- grid.12380.380000 0004 1754 9227Section Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1118, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Changes in the histopathology and in the proteins related to the MAPK pathway in the brains of rats exposed to pre and postnatal radiofrequency radiation over four generations. J Chem Neuroanat 2022; 126:102187. [DOI: 10.1016/j.jchemneu.2022.102187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 10/26/2022] [Accepted: 10/26/2022] [Indexed: 11/09/2022]
|
14
|
Rawat P, Sehar U, Bisht J, Selman A, Culberson J, Reddy PH. Phosphorylated Tau in Alzheimer's Disease and Other Tauopathies. Int J Mol Sci 2022; 23:12841. [PMID: 36361631 PMCID: PMC9654278 DOI: 10.3390/ijms232112841] [Citation(s) in RCA: 165] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/21/2022] [Accepted: 10/22/2022] [Indexed: 07/29/2023] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in elderly people. Amyloid beta (Aβ) deposits and neurofibrillary tangles are the major pathological features in an Alzheimer's brain. These proteins are highly expressed in nerve cells and found in most tissues. Tau primarily provides stabilization to microtubules in the part of axons and dendrites. However, tau in a pathological state becomes hyperphosphorylated, causing tau dysfunction and leading to synaptic impairment and degeneration of neurons. This article presents a summary of the role of tau, phosphorylated tau (p-tau) in AD, and other tauopathies. Tauopathies, including Pick's disease, frontotemporal dementia, corticobasal degeneration, Alzheimer's disease, argyrophilic grain disease, progressive supranuclear palsy, and Huntington's disease, are the result of misprocessing and accumulation of tau within the neuronal and glial cells. This article also focuses on current research on the post-translational modifications and genetics of tau, tau pathology, the role of tau in tauopathies and the development of new drugs targeting p-tau, and the therapeutics for treating and possibly preventing tauopathies.
Collapse
Affiliation(s)
- Priyanka Rawat
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Jasbir Bisht
- Department of Pediatrics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Ashley Selman
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - John Culberson
- Department of Family Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - P. Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
15
|
Gutierrez BA, Limon A. Synaptic Disruption by Soluble Oligomers in Patients with Alzheimer's and Parkinson's Disease. Biomedicines 2022; 10:1743. [PMID: 35885050 PMCID: PMC9313353 DOI: 10.3390/biomedicines10071743] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 01/10/2023] Open
Abstract
Neurodegenerative diseases are the result of progressive dysfunction of the neuronal activity and subsequent neuronal death. Currently, the most prevalent neurodegenerative diseases are by far Alzheimer's (AD) and Parkinson's (PD) disease, affecting millions of people worldwide. Although amyloid plaques and neurofibrillary tangles are the neuropathological hallmarks for AD and Lewy bodies (LB) are the hallmark for PD, current evidence strongly suggests that oligomers seeding the neuropathological hallmarks are more toxic and disease-relevant in both pathologies. The presence of small soluble oligomers is the common bond between AD and PD: amyloid β oligomers (AβOs) and Tau oligomers (TauOs) in AD and α-synuclein oligomers (αSynOs) in PD. Such oligomers appear to be particularly increased during the early pathological stages, targeting synapses at vulnerable brain regions leading to synaptic plasticity disruption, synapse loss, inflammation, excitation to inhibition imbalance and cognitive impairment. Absence of TauOs at synapses in individuals with strong AD disease pathology but preserved cognition suggests that mechanisms of resilience may be dependent on the interactions between soluble oligomers and their synaptic targets. In this review, we will discuss the current knowledge about the interactions between soluble oligomers and synaptic dysfunction in patients diagnosed with AD and PD, how it affects excitatory and inhibitory synaptic transmission, and the potential mechanisms of synaptic resilience in humans.
Collapse
Affiliation(s)
| | - Agenor Limon
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, School of Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA;
| |
Collapse
|
16
|
Pandey N, Vinod PK. Model scenarios for cell cycle re-entry in Alzheimer's disease. iScience 2022; 25:104543. [PMID: 35747391 PMCID: PMC9209725 DOI: 10.1016/j.isci.2022.104543] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 05/01/2022] [Accepted: 06/02/2022] [Indexed: 11/30/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease. Aberrant production and aggregation of amyloid beta (Aβ) peptide into plaques is a frequent feature of AD, but therapeutic approaches targeting Aβ accumulation fail to inhibit disease progression. The approved cholinesterase inhibitor drugs are symptomatic treatments. During human brain development, the progenitor cells differentiate into neurons and switch to a postmitotic state. However, cell cycle re-entry often precedes loss of neurons. We developed mathematical models of multiple routes leading to cell cycle re-entry in neurons that incorporate the crosstalk between cell cycle, neuronal, and apoptotic signaling mechanisms. We show that the integration of multiple feedback loops influences disease severity making the switch to pathological state irreversible. We observe that the transcriptional changes associated with this transition are also characteristics of the AD brain. We propose that targeting multiple arms of the feedback loop may bring about disease-modifying effects in AD. Developed mathematical models of cell cycle re-entry in Alzheimer's disease (AD) Integration of multiple feedback loops drives irreversible transition to AD Predicted transcriptional dysregulation is validated using AD gene expression data Inhibition of self-amplifying feedback loops brings about disease-modifying effects
Collapse
Affiliation(s)
- Nishtha Pandey
- Center for Computational Natural Sciences and Bioinformatics, International Institute of Information Technology, Hyderabad 500032 India
| | - P K Vinod
- Center for Computational Natural Sciences and Bioinformatics, International Institute of Information Technology, Hyderabad 500032 India
| |
Collapse
|
17
|
Incontri-Abraham D, Esparza-Salazar FJ, Ibarra A. Copolymer-1 as a potential therapy for mild cognitive impairment. Brain Cogn 2022; 162:105892. [PMID: 35841771 DOI: 10.1016/j.bandc.2022.105892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/28/2022] [Indexed: 11/25/2022]
Abstract
Mild cognitive impairment (MCI) is a prodromal stage of memory impairment that may precede dementia. MCI is classified by the presence or absence of memory impairment into amnestic or non-amnestic MCI, respectively. More than 90% of patients with amnestic MCI who progress towards dementia meet criteria for Alzheimer's disease (AD). A combination of mechanisms promotes MCI, including intracellular neurofibrillary tangle formation, extracellular amyloid deposition, oxidative stress, neuronal loss, synaptodegeneration, cholinergic dysfunction, cerebrovascular disease, and neuroinflammation. However, emerging evidence indicates that neuroinflammation plays an important role in the pathogenesis of cognitive impairment. Unfortunately, there are currently no Food and Drug Administration (FDA)-approved drugs for MCI. Copolymer-1 (Cop-1), also known as glatiramer acetate, is a synthetic polypeptide of four amino acids approved by the FDA for the treatment of relapsing-remitting multiple sclerosis. Cop-1 therapeutic effect is attributed to immunomodulation, promoting a switch from proinflammatory to anti-inflammatory phenotype. In addition to its anti-inflammatory properties, it stimulates brain-derived neurotrophic factor (BDNF) secretion, a neurotrophin involved in neurogenesis and the generation of hippocampal long-term potentials. Moreover, BDNF levels are significantly decreased in patients with cognitive impairment. Therefore, Cop-1 immunization might promote synaptic plasticity and memory consolidation by increasing BDNF production in patients with MCI.
Collapse
Affiliation(s)
- Diego Incontri-Abraham
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Av. Universidad Anáhuac No. 46, Col. Lomas Anáhuac, Huixquilucan, CP 52786, Edo. de México, Mexico
| | - Felipe J Esparza-Salazar
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Av. Universidad Anáhuac No. 46, Col. Lomas Anáhuac, Huixquilucan, CP 52786, Edo. de México, Mexico
| | - Antonio Ibarra
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, Av. Universidad Anáhuac No. 46, Col. Lomas Anáhuac, Huixquilucan, CP 52786, Edo. de México, Mexico.
| |
Collapse
|
18
|
Advancements in the development of multi-target directed ligands for the treatment of Alzheimer's disease. Bioorg Med Chem 2022; 61:116742. [PMID: 35398739 DOI: 10.1016/j.bmc.2022.116742] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/01/2022] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial irreversible neurological disorder which results in cognitive impairment, loss of cholinergic neurons in synapses of the basal forebrain and neuronal death. Exact pathology of the disease is not yet known however, many hypotheses have been proposed for its treatment. The available treatments including monotherapies and combination therapies are not able to combat the disease effectively because of its complex pathological mechanism. A multipotent drug for AD has the potential to bind or inhibit multiple targets responsible for the progression of the disease like aggregated Aβ, hyperphosphorylated tau proteins, cholinergic and adrenergic receptors, MAO enzymes, overactivated N-methyl-d-aspartate (NMDA), α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor etc. The traditional approach of one disease-one target-one drug has been rationalized to one drug-multi targets for the chronic diseases like AD and cancer. Thus, over the last decade research focus has been shifted towards the development of multi target directed ligands (MTDLs) which can simultaneously inhibit multiple targets and stop or slow the progression of the disease. The MTDLs can be more effective against AD and eliminate any possibility of drug-drug interactions. Many important active pharmacophore units have been fused, merged or incorporated into different scaffolds to synthesize new potent drugs. In the current article, we have described various hypothesis for AD and effectiveness of the MTDLs treatment strategy is discussed in detail. Different chemical scaffolds and their synthetic strategies have been described and important functionalities are identified in the chemical scaffold that have the potential to bind to the multiple targets. The important leads identified in this study with MTDL characteristics have the potential to be developed as drug candidates for the effective treatment of AD.
Collapse
|
19
|
van der Westhuizen CJ, Stander A, Riley DL, Panayides JL. Discovery of Novel Acetylcholinesterase Inhibitors by Virtual Screening, In Vitro Screening, and Molecular Dynamics Simulations. J Chem Inf Model 2022; 62:1550-1572. [PMID: 35139637 DOI: 10.1021/acs.jcim.1c01443] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Alzheimer's disease is the most common neurodegenerative disease and currently poses a significant socioeconomic problem. This study describes the uses of computer-aided drug discovery techniques to identify novel inhibitors of acetylcholinesterase, a target for Alzheimer's disease. High-throughput virtual screening was employed to predict potential inhibitors of acetylcholinesterase. Validation of enrichment was performed with the DUD-E data set, showing that an ensemble of binding pocket conformations is critical when a diverse set of ligands are being screened. A total of 720 compounds were submitted for in vitro screening, which led to 25 hits being identified with IC50 values of less than 50 μM. The majority of these hits belonged to two scaffolds: 1-ethyl-3-methoxy-3-methylpyrrolidine and 1H-pyrrolo[3,2-c]pyridin-6-amine both of which are noted to be promising compounds for further optimization. As various possible binding poses were suggested from molecular docking, molecular dynamics simulations were employed to validate the poses. In the case of the most active compounds identified, a critical, stable water bridge formed deep within the binding pocket was identified potentially explaining in part the lack of activity for subsets of compounds that are not able to form this water bridge.
Collapse
Affiliation(s)
- C Johan van der Westhuizen
- Department of Chemistry, Faculty of Natural and Agricultural Sciences, University of Pretoria, Lynnwood Road, Pretoria 0028, South Africa.,Pharmaceutical Technologies, CSIR Future Production: Chemicals, Meiring Naudé Road, Pretoria 0184, South Africa
| | - André Stander
- Department of Physiology, Faculty of Health Science, University of Pretoria, Lynnwood Road, Pretoria 0031, South Africa
| | - Darren L Riley
- Department of Chemistry, Faculty of Natural and Agricultural Sciences, University of Pretoria, Lynnwood Road, Pretoria 0028, South Africa
| | - Jenny-Lee Panayides
- Pharmaceutical Technologies, CSIR Future Production: Chemicals, Meiring Naudé Road, Pretoria 0184, South Africa
| |
Collapse
|
20
|
Tay EXY, Chia K, Ong DST. Epigenetic plasticity and redox regulation of neural stem cell state and fate. Free Radic Biol Med 2021; 170:116-130. [PMID: 33684459 DOI: 10.1016/j.freeradbiomed.2021.02.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 01/20/2021] [Accepted: 02/24/2021] [Indexed: 12/16/2022]
Abstract
The neural stem cells (NSCs) are essential for normal brain development and homeostasis. The cell state (i.e. quiescent versus activated) and fate (i.e. the cell lineage of choice upon differentiation) of NSCs are tightly controlled by various redox and epigenetic regulatory mechanisms. There is an increasing appreciation that redox and epigenetic regulations are intimately linked, but how this redox-epigenetics crosstalk affects NSC activity remains poorly understood. Another unresolved topic is whether the NSCs actually contribute to brain ageing and neurodegenerative diseases. In this review, we aim to 1) distill concepts that underlie redox and epigenetic regulation of NSC state and fate; 2) provide examples of the redox-epigenetics crosstalk in NSC biology; and 3) highlight potential redox- and epigenetic-based therapeutic opportunities to rescue NSC dysfunctions in ageing and neurodegenerative diseases.
Collapse
Affiliation(s)
- Emmy Xue Yun Tay
- Department of Physiology, National University of Singapore, Singapore, 117593, Singapore
| | - Kimberly Chia
- Department of Physiology, National University of Singapore, Singapore, 117593, Singapore
| | - Derrick Sek Tong Ong
- Department of Physiology, National University of Singapore, Singapore, 117593, Singapore; Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore; National Neuroscience Institute, Singapore, 308433, Singapore.
| |
Collapse
|
21
|
Balendra V, Singh SK. Therapeutic potential of astaxanthin and superoxide dismutase in Alzheimer's disease. Open Biol 2021; 11:210013. [PMID: 34186009 PMCID: PMC8241491 DOI: 10.1098/rsob.210013] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress, the imbalance of the antioxidant system, results in an accumulation of neurotoxic proteins in Alzheimer's disease (AD). The antioxidant system is composed of exogenous and endogenous antioxidants to maintain homeostasis. Superoxide dismutase (SOD) is an endogenous enzymatic antioxidant that converts superoxide ions to hydrogen peroxide in cells. SOD supplementation in mice prevented cognitive decline in stress-induced cells by reducing lipid peroxidation and maintaining neurogenesis in the hippocampus. Furthermore, SOD decreased expression of BACE1 while reducing plaque burden in the brain. Additionally, Astaxanthin (AST), a potent exogenous carotenoid, scavenges superoxide anion radicals. Mice treated with AST showed slower memory decline and decreased depositions of amyloid-beta (Aβ) and tau protein. Currently, the neuroprotective potential of these supplements has only been examined separately in studies. However, a single antioxidant cannot sufficiently resist oxidative damage to the brain, therefore, a combinatory approach is proposed as a relevant therapy for ameliorating pathological changes in AD.
Collapse
Affiliation(s)
| | - Sandeep Kumar Singh
- Indian Scientific Education and Technology (ISET) Foundation, Lucknow 226002, India
| |
Collapse
|
22
|
Wang YR, Chuang HC, Tripathi A, Wang YL, Ko ML, Chuang CC, Chen JC. High-Sensitivity and Trace-Amount Specimen Electrochemical Sensors for Exploring the Levels of β-Amyloid in Human Blood and Tears. Anal Chem 2021; 93:8099-8106. [PMID: 34047190 DOI: 10.1021/acs.analchem.0c04980] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
As the occurrence of Alzheimer's disease (AD) has increased, the detection and treatment of AD have become global social issues. Effective early detection and wide-range screening of AD allow patients to gain early control and delay brain degeneration. For these reasons, we choose electrochemical sensors to complete the detection task. Although bio-electrochemical technology for antibody and antigen sensing is not a new technology, considering the scarcity of tear samples for dementia and since the existing AD detection techniques are highly invasive and expensive for subjects, we have to use the traditional detection techniques for the early screening of Alzheimer's disease via trace-amount specimens. An AD-related protein in the eye is thought to be an important biomarker for early detection. To carry out detection using tear samples as a test specimens, a tear collection device was developed in this study that extracted 10 μL of tear fluid from a tear Schirmer strip. In this research, we distinguished healthy people in different age groups and detect Aβ in both tear and blood samples. We developed a biosensor, which could detect Aβ in tear specimen from 1 to 100 pg/mL. Also, this biosensor is inexpensive, disposable, and easy to use. In our result, the concentration of Aβ in tears was approximately 10 times more than that in blood. This study demonstrates the feasibility and prospects of future screening for AD-associated biomarkers by a dynamic comparison between blood and tears.
Collapse
Affiliation(s)
- Yu-Rong Wang
- Institute of Biomedical Engineering, National Chiao Tung University, Hsinchu 30010, Taiwan
| | - Han-Chien Chuang
- Institute of Biomedical Engineering, National Chiao Tung University, Hsinchu 30010, Taiwan
| | - Adarsh Tripathi
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30010, Taiwan.,Institute of Nanoengineering and Microsystems, National Tsing Hua University, Hsinchu 30010, Taiwan
| | - Yu-Lin Wang
- Institute of Nanoengineering and Microsystems, National Tsing Hua University, Hsinchu 30010, Taiwan.,Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu 30010, Taiwan
| | - Mei-Lan Ko
- Department of Ophthalmology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu 30010, Taiwan.,Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 30010, Taiwan
| | - Ching-Cheng Chuang
- Institute of Biomedical Engineering, National Chiao Tung University, Hsinchu 30010, Taiwan.,Department of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 30010, Taiwan
| | - Jung-Chih Chen
- Institute of Biomedical Engineering, National Chiao Tung University, Hsinchu 30010, Taiwan.,Department of Electrical and Computer Engineering, National Chiao Tung University, Hsinchu 30010, Taiwan.,Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 30010, Taiwan
| |
Collapse
|
23
|
Sordo L, Martini AC, Houston EF, Head E, Gunn-Moore D. Neuropathology of Aging in Cats and its Similarities to Human Alzheimer’s Disease. FRONTIERS IN AGING 2021; 2:684607. [PMID: 35822024 PMCID: PMC9261448 DOI: 10.3389/fragi.2021.684607] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/27/2021] [Indexed: 11/15/2022]
Abstract
Elderly cats develop age-related behavioral and neuropathological changes that ultimately lead to cognitive dysfunction syndrome (CDS). These neuropathologies share similarities to those seen in the brains of humans with Alzheimer’s disease (AD), including the extracellular accumulation of ß-amyloid (Aβ) and intraneuronal deposits of hyperphosphorylated tau, which are considered to be the two major hallmarks of AD. The present study assessed the presence and distribution of Aβ and tau hyperphosphorylation within the cat brain (n = 55 cats), and how the distribution of these proteins changes with age and the presence of CDS. For this, immunohistochemistry was performed on seven brain regions from cats of various ages, with and without CDS (n = 10 with CDS). Cats accumulate both intracytoplasmic and extracellular deposits of Aβ, as well as intranuclear and intracytoplasmic hyperphosphorylated tau deposits. Large extracellular aggregates of Aβ were found in elderly cats, mainly in the cortical brain areas, with occasional hippocampal aggregates. This may suggest that these aggregates start in cortical areas and later progress to the hippocampus. While Aβ senile plaques in people with AD have a dense core, extracellular Aβ deposits in cats exhibited a diffuse pattern, similar to the early stages of plaque pathogenesis. Intraneuronal Aβ deposits were also observed, occurring predominantly in cortical brain regions of younger cats, while older cats had few to no intraneuronal Aβ deposits, especially when extracellular aggregates were abundant. Intracytoplasmic hyperphosphorylated tau was found within neurons in the brains of elderly cats, particularly in those with CDS. Due to their ultrastructural features, these deposits are considered to be pre-tangles, which are an early stage of the neurofibrillary tangles seen in AD. The largest numbers of pre-tangles are found mainly in the cerebral cortex of elderly cats, whereas lower numbers were found in other regions (i.e., entorhinal cortex and hippocampus). For the first time, intranuclear tau was found in both phosphorylated and non-phosphorylated states within neurons in the cat brain. The highest numbers of intranuclear deposits were found in the cortex of younger cats, and this tended to decrease with age. In contrast, elderly cats with pre-tangles had only occasional or no nuclear labelling.
Collapse
Affiliation(s)
- Lorena Sordo
- The Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
- *Correspondence: Lorena Sordo,
| | - Alessandra C. Martini
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, United States
| | - E. Fiona Houston
- The Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, United States
| | - Danièlle Gunn-Moore
- The Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
24
|
Saleem U, Hira S, Anwar F, Shah MA, Bashir S, Baty RS, Badr RH, Blundell R, Batiha GES, Ahmad B. Pharmacological Screening of Viola odorata L . for Memory-Enhancing Effect via Modulation of Oxidative Stress and Inflammatory Biomarkers. Front Pharmacol 2021; 12:664832. [PMID: 34149418 PMCID: PMC8210412 DOI: 10.3389/fphar.2021.664832] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/22/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose: Alzheimer disease (AD) is a progressive neurodegenerative disorder that is caused by neuroinflammation and oxidative stress. The present study aimed to characterize and then investigate the memory-enhancing potential of Viola odorata methanolic extract in lipopolysaccharide (LPS)–treated mice. Methods:V. odorata characterization was done by using the GCMS technique. Neuroinflammation was induced by the intracerebroventricular administration of LPS at a dose of 12 µg. Animals were divided randomly into six groups (n = 10). Group I was normal control, which was given vehicle. Group II was disease control, which received LPS (12 µg) via the intracerebroventricular route. Group III was standard, which was administered with donepezil (3 µg) orally for 21 days. Groups IV–VI were the treatment groups, which were administered with the extract at 100, 200, and 400 mg/kg dose levels orally respectively for 21 days. Groups III–VI received LPS (12 µg) on the first day along with their treatments. During the treatment, the animals were assessed for memory retention by employing different behavioral paradigms namely elevated plus maze, passive avoidance, foot shock and open field. Various mediators [endogenous antioxidants, neurotransmitters, and acetylcholinesterase (AChE)] involved in the pathogenesis of AD were quantified by using the UV spectrophotometric method. Results: Extract-treated groups showed a remarkable improvement in cognitive impairment in all behavioral paradigms. Oxidative stress biomarkers, that is, superoxide dismutase, catalase, and glutathione were raised dose-dependently in the treatment groups with a dose-dependent decrease in the malonaldehyde and AChE levels in the brains of the treated animals. The treatment groups showed decreased levels of inflammatory biomarkers, that is, tumor necrosis factor–alpha, nuclear factor kappa light-chain enhancer of activated β-cells, and cyclo-oxygenase, which supports the therapeutic effectiveness of the treatment. Conclusion: Based on behavioral, oxidative stress biomarker, and neuroinflammatory data, it is concluded that V. odorata possesses memory-enhancing activity and may prove a beneficial role in the management of AD.
Collapse
Affiliation(s)
- Uzma Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Sundas Hira
- Riphah Institute of Pharmaceutical Sciences, Riphah International University Lahore, Lahore, Pakistan
| | - Fareeha Anwar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University Lahore, Lahore, Pakistan
| | - Muhammad Ajmal Shah
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Samia Bashir
- Riphah Institute of Pharmaceutical Sciences, Riphah International University Lahore, Lahore, Pakistan
| | - Roua S Baty
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
| | - Reem H Badr
- Department of Plant Physiology Botany and Microbiology, Faculty of Science, Alex University, Alexandria, Egypt
| | - Renald Blundell
- American University of Malta, Triq Dom Mintoff, Bormla, Malta
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Bashir Ahmad
- Riphah Institute of Pharmaceutical Sciences, Riphah International University Lahore, Lahore, Pakistan
| |
Collapse
|
25
|
Preclinical Marmoset Model for Targeting Chronic Inflammation as a Strategy to Prevent Alzheimer's Disease. Vaccines (Basel) 2021; 9:vaccines9040388. [PMID: 33920929 PMCID: PMC8071309 DOI: 10.3390/vaccines9040388] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/08/2021] [Accepted: 04/13/2021] [Indexed: 11/17/2022] Open
Abstract
Due to the aging population, modern society is facing an increasing prevalence of neurological diseases such as Alzheimer’s disease (AD). AD is an age-related chronic neurodegenerative disorder for which no satisfying therapy exists. Understanding the mechanisms underlying the onset of AD is necessary to find targets for protective treatment. There is growing awareness of the essential role of the immune system in the early AD pathology. Amyloidopathy, the main feature of early-stage AD, has a deregulating effect on the immune function. This is reciprocal as the immune system also affects amyloidopathy. It seems that the inflammatory reaction shows a heterogeneous pattern depending on the stage of the disease and the variation between individuals, making not only the target but also the timing of treatment important. The lack of relevant translational animal models that faithfully reproduce clinical and pathogenic features of AD is a major cause of the delay in developing new disease-modifying therapies and their optimal timing of administration. This review describes the communication between amyloidopathy and inflammation and the possibility of using nonhuman primates as a relevant animal model for preclinical AD research.
Collapse
|
26
|
Gil L, Niño SA, Capdeville G, Jiménez-Capdeville ME. Aging and Alzheimer's disease connection: Nuclear Tau and lamin A. Neurosci Lett 2021; 749:135741. [PMID: 33610669 DOI: 10.1016/j.neulet.2021.135741] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 01/12/2021] [Accepted: 02/11/2021] [Indexed: 12/24/2022]
Abstract
Age-related pathologies like Alzheimer`s disease (AD) imply cellular responses directed towards repairing DNA damage. Postmitotic neurons show progressive accumulation of oxidized DNA during decades of brain aging, which is especially remarkable in AD brains. The characteristic cytoskeletal pathology of AD neurons is brought about by the progressive changes that neurons undergo throughout aging, and their irreversible nuclear transformation initiates the disease. This review focusses on critical molecular events leading to the loss of plasticity that underlies cognitive deficits in AD. During healthy neuronal aging, nuclear Tau participates in the regulation of the structure and function of the chromatin. The aberrant cell cycle reentry initiated for DNA repair triggers a cascade of events leading to the dysfunctional AD neuron, whereby Tau protein exits the nucleus leading to chromatin disorganization. Lamin A, which is not typically expressed in neurons, appears at the transformation from senile to AD neurons and contributes to halting the consequences of cell cycle reentry and nuclear Tau exit, allowing the survival of the neuron. Nevertheless, this irreversible nuclear transformation alters the nucleic acid and protein synthesis machinery as well as the nuclear lamina and cytoskeleton structures, leading to neurofibrillary tangles formation and final neurodegeneration.
Collapse
Affiliation(s)
- Laura Gil
- Departamento de Genética, Escuela de Medicina, Universidad "Alfonso X el Sabio", Madrid, Spain
| | - Sandra A Niño
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, Mexico
| | | | | |
Collapse
|
27
|
Colnaghi L, Rondelli D, Muzi-Falconi M, Sertic S. Tau and DNA Damage in Neurodegeneration. Brain Sci 2020; 10:E946. [PMID: 33297375 PMCID: PMC7762255 DOI: 10.3390/brainsci10120946] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/29/2020] [Accepted: 12/02/2020] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative disorders are a family of incurable conditions. Among them, Alzheimer's disease and tauopathies are the most common. Pathological features of these two disorders are synaptic loss, neuronal cell death and increased DNA damage. A key pathological protein for the onset and progression of the conditions is the protein tau, a microtubule-binding protein highly expressed in neurons and encoded by the MAPT (microtubule-associated protein tau) gene. Tau is predominantly a cytosolic protein that interacts with numerous other proteins and molecules. Recent findings, however, have highlighted new and unexpected roles for tau in the nucleus of neuronal cells. This review summarizes the functions of tau in the metabolism of DNA, describing them in the context of the disorders.
Collapse
Affiliation(s)
- Luca Colnaghi
- Department of Molecular Biochemistry and Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Negri 2, 20156 Milan, Italy
| | - Diego Rondelli
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy; (D.R.); (M.M.-F.)
| | - Marco Muzi-Falconi
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy; (D.R.); (M.M.-F.)
| | - Sarah Sertic
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy; (D.R.); (M.M.-F.)
| |
Collapse
|
28
|
Neurodegenerative Implications of Neuronal Cytoplasmic Protein Dysfunction in Response to Environmental Contaminants. Neurotox Res 2020; 39:533-541. [PMID: 33175324 DOI: 10.1007/s12640-020-00308-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/10/2020] [Accepted: 10/29/2020] [Indexed: 10/23/2022]
Abstract
Neurodegenerative diseases account for a significant portion of public health concerns particularly in the aging population. The dysfunction of interfilament proteins has been identified as a key event in the initiation of neurodegeneration and subsequent progression to neurodegenerative diseases. In addition, several studies have found associations between the dysfunction of interfilament proteins and exposure to environmental contaminants. Therefore, in this review, the role of interfilament proteins in neuronal cells, their connection to neurotoxicity from environmental contaminants, and finally the resulting neurodegeneration are discussed.
Collapse
|
29
|
Vegh C, Stokes K, Ma D, Wear D, Cohen J, Ray SD, Pandey S. A Bird's-Eye View of the Multiple Biochemical Mechanisms that Propel Pathology of Alzheimer's Disease: Recent Advances and Mechanistic Perspectives on How to Halt the Disease Progression Targeting Multiple Pathways. J Alzheimers Dis 2020; 69:631-649. [PMID: 31127770 PMCID: PMC6598003 DOI: 10.3233/jad-181230] [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] [Indexed: 12/12/2022]
Abstract
Neurons consume the highest amount of oxygen, depend on oxidative metabolism for energy, and survive for the lifetime of an individual. Therefore, neurons are vulnerable to death caused by oxidative-stress, accumulation of damaged and dysfunctional proteins and organelles. There is an exponential increase in the number of patients diagnosed with neurodegenerative diseases such as Alzheimer's (AD) as the number of elderly increases exponentially. Development of AD pathology is a complex phenomenon characterized by neuronal death, accumulation of extracellular amyloid-β plaques and neurofibrillary tangles, and most importantly loss of memory and cognition. These pathologies are most likely caused by mechanisms including oxidative stress, mitochondrial dysfunction/stress, accumulation of misfolded proteins, and defective organelles due to impaired proteasome and autophagy mechanisms. Currently, there are no effective treatments to halt the progression of this disease. In order to treat this complex disease with multiple biochemical pathways involved, a complex treatment regimen targeting different mechanisms should be investigated. Furthermore, as AD is a progressive disease-causing morbidity over many years, any chemo-modulator for treatment must be used over long period of time. Therefore, treatments must be safe and non-interfering with other processes. Ideally, a treatment like medicinal food or a supplement that can be taken regularly without any side effect capable of reducing oxidative stress, stabilizing mitochondria, activating autophagy or proteasome, and increasing energy levels of neurons would be the best solution. This review summarizes progress in research on different mechanisms of AD development and some of the potential therapeutic development strategies targeting the aforementioned pathologies.
Collapse
Affiliation(s)
- Caleb Vegh
- Department of Chemistry and Biochemistry University of Windsor, Ontario, Canada
| | - Kyle Stokes
- Department of Chemistry and Biochemistry University of Windsor, Ontario, Canada
| | - Dennis Ma
- Department of Chemistry and Biochemistry University of Windsor, Ontario, Canada
| | - Darcy Wear
- Department of Chemistry and Biochemistry University of Windsor, Ontario, Canada
| | - Jerome Cohen
- Department of Psychology University of Windsor, Ontario, Canada
| | - Sidhartha D Ray
- Department of Pharmaceutical and Biomedical Sciences, Touro College of Pharmacy and School of Medicine, Manhattan, NY, USA
| | - Siyaram Pandey
- Department of Chemistry and Biochemistry University of Windsor, Ontario, Canada
| |
Collapse
|
30
|
Li J, Wang G, Qin Y, Zhang X, Wang HF, Liu HW, Zhu LJ, Yao XS. Neuroprotective constituents from the aerial parts of Cannabis sativa L. subsp. sativa. RSC Adv 2020; 10:32043-32049. [PMID: 35518127 PMCID: PMC9056577 DOI: 10.1039/d0ra04565a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 08/12/2020] [Indexed: 11/21/2022] Open
Abstract
Five new compounds including three new cannabinoids, cannabisativas A-C (1-3), two new phenolic acids, (7Z,9Z)-cannabiphenolic acid A (4) and (8S,9Z)-cannabiphenolic acid B (5), together with twelve known compounds (6-17), were isolated from the aerial parts of Cannabis sativa L. subsp. sativa. The structures of 1-5 were established on the basis of extensive 1D, 2D NMR and HRESIMS analysis. The absolute configurations were determined by comparison between their experimental and calculated spectra of electronic circular dichroism (ECD) or the modified Mosher's method. The neuroprotective effects of the compounds 1-17 were evaluated on PC 12 cells. Compounds 12, 13 and 15 showed potential protective effects against H2O2-induced damage.
Collapse
Affiliation(s)
- Jia Li
- College of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University Shenyang 110016 China
| | - Guan Wang
- College of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University Shenyang 110016 China
- State Key Laboratory of Biotherapy, Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| | - Yu Qin
- College of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University Shenyang 110016 China
| | - Xue Zhang
- College of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University Shenyang 110016 China
| | - Hai-Feng Wang
- College of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University Shenyang 110016 China
| | - Hong-Wei Liu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences Beijing 100101 China
| | - Ling-Juan Zhu
- College of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University Shenyang 110016 China
- State Key Laboratory of Biotherapy, Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| | - Xin-Sheng Yao
- College of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University Shenyang 110016 China
| |
Collapse
|
31
|
Muralidar S, Ambi SV, Sekaran S, Thirumalai D, Palaniappan B. Role of tau protein in Alzheimer's disease: The prime pathological player. Int J Biol Macromol 2020; 163:1599-1617. [PMID: 32784025 DOI: 10.1016/j.ijbiomac.2020.07.327] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 07/07/2020] [Accepted: 07/31/2020] [Indexed: 01/11/2023]
Abstract
Alzheimer's disease (AD) is a prevalently found tauopathy characterized by memory loss and cognitive insufficiency. AD is an age-related neurodegenerative disease with two major hallmarks which includes extracellular amyloid plaques made of amyloid-β (Aβ) and intracellular neurofibrillary tangles of hyperphosphorylated tau. With population aging worldwide, there is an indispensable need for treatment strategies that can potentially manage this developing dementia. Despite broad researches on targeting Aβ in the past two decades, research findings on Aβ targeted therapeutics failed to prove efficacy in the treatment of AD. Tau protein with its extensive pathological role in several neurodegenerative diseases can be considered as a promising target candidate for developing therapeutic interventions. The abnormal hyperphosphorylation of tau plays detrimental pathological functions which ultimately lead to neurodegeneration. This review will divulge the importance of tau in AD pathogenesis, the interplay of Aβ and tau, the pathological functions of tau, and potential therapeutic strategies for an effective management of neuronal disorders.
Collapse
Affiliation(s)
- Shibi Muralidar
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India
| | - Senthil Visaga Ambi
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India.
| | - Saravanan Sekaran
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India; Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India
| | - Diraviyam Thirumalai
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India
| | - Balamurugan Palaniappan
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India
| |
Collapse
|
32
|
Kabir MT, Sufian MA, Uddin MS, Begum MM, Akhter S, Islam A, Mathew B, Islam MS, Amran MS, Md Ashraf G. NMDA Receptor Antagonists: Repositioning of Memantine as a Multitargeting Agent for Alzheimer's Therapy. Curr Pharm Des 2020; 25:3506-3518. [PMID: 31604413 DOI: 10.2174/1381612825666191011102444] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 10/08/2019] [Indexed: 12/28/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that causes problems with memory, thinking, and behavior. Currently, there is no drug that can reduce the pathological events of this degenerative disease but symptomatic relief is possible that can abate the disease condition. N-methyl-D-aspartate (NMDA) receptors exert a critical role for synaptic plasticity as well as transmission. Overstimulation of glutamate receptors, predominantly NMDA type, may cause excitotoxic effects on neurons and is recommended as a mechanism for neurodegeneration. Atypical activation of the NMDA receptor has been suggested for AD by synaptic dysfunction. NMDA receptor antagonists especially memantine block the NMDA receptor and can reduce the influx of calcium (Ca2+) ions into neuron, thus, toxic intracellular events are not activated. This review represents the role of NMDA receptors antagonists as potential therapeutic agents to reduce AD. Moreover, this review highlights the repositioning of memantine as a potential novel therapeutic multitargeting agent for AD.
Collapse
Affiliation(s)
| | | | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh.,Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | | | - Shammi Akhter
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | - Ariful Islam
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, United States
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | | | - Md Shah Amran
- Department of Pharmaceutical Chemistry, University of Dhaka, Dhaka, Bangladesh
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
33
|
Pakdeepak K, Chokchaisiri R, Tocharus J, Jearjaroen P, Tocharus C, Suksamrarn A. 5,6,7,4'-Tetramethoxyflavanone protects against neuronal degeneration induced by dexamethasone by attenuating amyloidogenesis in mice. EXCLI JOURNAL 2020; 19:16-32. [PMID: 32038114 PMCID: PMC7003641 DOI: 10.17179/excli2019-1940] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/04/2019] [Indexed: 12/31/2022]
Abstract
Long-term exposure to high glucocorticoid levels induces memory impairment and neurodegeneration in Alzheimer's disease (AD) by increasing the expression of amyloid β and tau hyperphosphorylation (pTau). Previous studies showed beneficial effects of flavonoids in neurodegenerative models. 5,6,7,4'-tetramethoxyflavanone (TMF) is one of the active ingredients in Chromolaena odorata (L.), which R. M. King and H. Rob discovered in Thailand. This study focused on the effects of TMF on dexamethasone (DEX)-induced neurodegeneration, amyloidogenesis, pTau expression, neuron synaptic function, and cognitive impairment and the potential mechanisms involved. Mice were intraperitoneally administered DEX for 28 days before being treated with TMF for 30 days. The mice were randomly divided into six groups (twelve mice per group): control; TMF administration (40 mg/kg); pioglitazone administration (20 mg/kg); DEX administration (60 mg/kg); DEX administration plus TMF; and DEX administration plus pioglitazone. Behavioral tests showed that TMF significantly attenuated the memory impairment triggered by DEX. Consistently, TMF reduced DEX-induced amyloid beta production by reducing the expression of beta-site APP cleaving enzyme 1 (BACE1) and presenilin 1 (PS1), whereas it increased the gene expression of a disintegrin and metalloprotease 10 (ADAM10). TMF treatment also decreased pTau expression, inhibited phosphonuclear factor-kappa B (pNF-kB) and inhibited glycogen synthase kinase 3 (GSK-3) activity by increasing GSK3 phosphorylation (pGSK3). In addition, TMF also improved synaptic function by increasing the expression of synaptophysin (Syn) and postsynaptic density protein 95 (PSD95) while decreasing acetylcholine esterase activity. Conclusively, TMF provided neuroprotection against DEX-induced neurodegeneration. These findings suggest that TMF might have potential as a therapeutic drug for AD.
Collapse
Affiliation(s)
- Kanet Pakdeepak
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.,Graduate School, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Jiraporn Tocharus
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Chainarong Tocharus
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Apichart Suksamrarn
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok 10240, Thailand
| |
Collapse
|
34
|
Carneiro P, Morais S, do Carmo Pereira M. Biosensors on the road to early diagnostic and surveillance of Alzheimer's disease. Talanta 2020; 211:120700. [PMID: 32070618 DOI: 10.1016/j.talanta.2019.120700] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/21/2019] [Accepted: 12/28/2019] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease is a debilitating and largely untreatable condition with subtle onset and slow progression over an extensive period of time, which culminate in increasing levels of disability. As Alzheimer's disease prevalence is expected to grow exponentially in the upcoming decades, there is an urgency to develop analytical technologies for the sensitive, reliable and cost-effective detection of Alzheimer's disease biomarkers. Biosensors are powerful analytical devices that translate events of biological recognition on physical or chemical transducers into electrical, thermal or optical signals. The high sensitivity and selectivity of biosensors associated with easy, rapid and low-cost determination of analytes have made this discipline one of the most intensively studied in the past decades. This review centers on recent advances, challenges and trends of Alzheimer's disease biosensing particularly in the effort to combine the unique properties of nanomaterials with biorecognition elements. In the last decade, impressive progresses have been made towards the development of biosensors, mainly electrochemical and optical, for detection of Alzheimer's disease biomarkers in the pico- and femto-molar range. Nonetheless, advances in multiplexed detection, robustness, stability and specificity are still necessary to ensure an accurate and differentiated diagnosis of this disease.
Collapse
Affiliation(s)
- Pedro Carneiro
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal; REQUIMTE-LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, R. Dr. António Bernardino de Almeida 431, 4200-072, Porto, Portugal
| | - Simone Morais
- REQUIMTE-LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, R. Dr. António Bernardino de Almeida 431, 4200-072, Porto, Portugal.
| | - Maria do Carmo Pereira
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465, Porto, Portugal
| |
Collapse
|
35
|
Srivastava A, Das B, Yao AY, Yan R. Metabotropic Glutamate Receptors in Alzheimer's Disease Synaptic Dysfunction: Therapeutic Opportunities and Hope for the Future. J Alzheimers Dis 2020; 78:1345-1361. [PMID: 33325389 PMCID: PMC8439550 DOI: 10.3233/jad-201146] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the presence of neuritic plaques and neurofibrillary tangles. The impaired synaptic plasticity and dendritic loss at the synaptic level is an early event associated with the AD pathogenesis. The abnormal accumulation of soluble oligomeric amyloid-β (Aβ), the major toxic component in amyloid plaques, is viewed to trigger synaptic dysfunctions through binding to several presynaptic and postsynaptic partners and thus to disrupt synaptic transmission. Over time, the abnormalities in neural transmission will result in cognitive deficits, which are commonly manifested as memory loss in AD patients. Synaptic plasticity is regulated through glutamate transmission, which is mediated by various glutamate receptors. Here we review recent progresses in the study of metabotropic glutamate receptors (mGluRs) in AD cognition. We will discuss the role of mGluRs in synaptic plasticity and their modulation as a possible strategy for AD cognitive improvement.
Collapse
Affiliation(s)
- Akriti Srivastava
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Brati Das
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Annie Y. Yao
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Riqiang Yan
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| |
Collapse
|
36
|
Mayaki AM, Abdul Razak IS, Noraniza MA, Mazlina M, Rasedee A. Biofluid Markers of Equine Neurological Disorders Reviewed From Human Perspectives. J Equine Vet Sci 2019; 86:102907. [PMID: 32067661 DOI: 10.1016/j.jevs.2019.102907] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 12/26/2019] [Accepted: 12/26/2019] [Indexed: 02/07/2023]
Abstract
Neurological disorders (NDs) are often fatal to horses. Thus, symptoms of equine NDs commonly indicate euthanasia. Current diagnostic approaches for equine NDs is based on clinical signs, differential diagnoses, analysis of cerebrospinal fluid (CSF), assessment of histopathological lesions, and imaging. However, advances in biofluid biomarkers in the diagnosis of human neurological diseases can potentially be applied to equine NDs. In this review, we described the established human blood and CSF neurobiomarkers that could potentially be used to diagnose equine NDs.
Collapse
Affiliation(s)
- Abubakar Musa Mayaki
- Department of Veterinary Pre-Clinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Intan Shameha Abdul Razak
- Department of Veterinary Pre-Clinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor, Malaysia.
| | - Mohd Adzahan Noraniza
- Department of Farm and Exotic Animal Medicine and Surgery, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Mazlan Mazlina
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Abdullah Rasedee
- Department of Veterinary Laboratory Diagnosis, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
37
|
Sabharwal V, Koushika SP. Crowd Control: Effects of Physical Crowding on Cargo Movement in Healthy and Diseased Neurons. Front Cell Neurosci 2019; 13:470. [PMID: 31708745 PMCID: PMC6823667 DOI: 10.3389/fncel.2019.00470] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 10/02/2019] [Indexed: 01/22/2023] Open
Abstract
High concentration of cytoskeletal filaments, organelles, and proteins along with the space constraints due to the axon's narrow geometry lead inevitably to intracellular physical crowding along the axon of a neuron. Local cargo movement is essential for maintaining steady cargo transport in the axon, and this may be impeded by physical crowding. Molecular motors that mediate active transport share movement mechanisms that allow them to bypass physical crowding present on microtubule tracks. Many neurodegenerative diseases, irrespective of how they are initiated, show increased physical crowding owing to the greater number of stalled organelles and structural changes associated with the cytoskeleton. Increased physical crowding may be a significant factor in slowing cargo transport to synapses, contributing to disease progression and culminating in the dying back of the neuronal process. This review explores the idea that physical crowding can impede cargo movement along the neuronal process. We examine the sources of physical crowding and strategies used by molecular motors that might enable cargo to circumvent physically crowded locations. Finally, we describe sub-cellular changes in neurodegenerative diseases that may alter physical crowding and discuss the implications of such changes on cargo movement.
Collapse
Affiliation(s)
| | - Sandhya P. Koushika
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| |
Collapse
|
38
|
Vyas S, Kothari S, Kachhwaha S. Nootropic medicinal plants: Therapeutic alternatives for Alzheimer’s disease. J Herb Med 2019. [DOI: 10.1016/j.hermed.2019.100291] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
39
|
Corriveau-Lecavalier N, Mellah S, Clément F, Belleville S. Evidence of parietal hyperactivation in individuals with mild cognitive impairment who progressed to dementia: A longitudinal fMRI study. Neuroimage Clin 2019; 24:101958. [PMID: 31357150 PMCID: PMC6664199 DOI: 10.1016/j.nicl.2019.101958] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 06/25/2019] [Accepted: 07/20/2019] [Indexed: 01/14/2023]
Abstract
Hyperactivation, which is defined as a higher level of activation in patients compared to cognitively unimpaired older adults (controls; CTL), might represent an early signature of Alzheimer's Disease (AD). The goal of this study was to assess the presence and location of hyperactivation in individuals with mild cognitive impairment (MCI) who were later diagnosed with dementia, examine how hyperactivation changes longitudinally, and whether it is related to time before dementia. Forty participants, 26 with MCI and 14 CTL were enrolled in the study. Magnetic resonance imaging was used to measure functional activation while participants encoded word-pairs as well as cortical thickness and regional brain volume at study entry (Y0) and two years later (Y2). Clinical follow-up was completed every two years following study entry to identify progressors (pMCI), that is, individuals who later received a diagnosis of dementia. Task-related activation was assessed in pMCI in both hippocampi and in regions showing greater cortical thinning from Y0 to Y2 compared to CTLs. Hyperactivation was found in pMCI individuals in the right supramarginal gyrus. Persons with pMCI also showed hypoactivation in the left hippocampus and left pars opercularis. Both hyper- and hypoactivation were present at Y0 and Y2 and did not change longitudinally. Activation was not associated with time before dementia diagnosis. Smaller volume and thinner cortical thickness were associated with shorter time to diagnosis in the left hippocampus and left pars opercularis. In conclusion, hyperactivation was found in individuals who later progressed to dementia, confirming that it might represent an early biomarker to identify individuals in the prodromal phase of AD and that its understanding could contribute to elucidate the key brain mechanisms that precede dementia.
Collapse
Affiliation(s)
- Nick Corriveau-Lecavalier
- Research Centre, Institut universitaire de gériatrie de Montréal, Canada; Department of Psychology, University of Montreal, Montreal, Canada
| | - Samira Mellah
- Research Centre, Institut universitaire de gériatrie de Montréal, Canada
| | - Francis Clément
- Research Centre, Institut universitaire de gériatrie de Montréal, Canada; Department of Psychology, University of Montreal, Montreal, Canada
| | - Sylvie Belleville
- Research Centre, Institut universitaire de gériatrie de Montréal, Canada; Department of Psychology, University of Montreal, Montreal, Canada.
| |
Collapse
|
40
|
Naserkhaki R, Zamanzadeh S, Baharvand H, Nabavi SM, Pakdaman H, Shahbazi S, Vosough M, Ghaedi G, Barzegar A, Mirtorabi D, Hedayatshodeh M, Ehsani E, Falahati M, Hajipour MJ, Shahpasand K. cis pT231-Tau Drives Neurodegeneration in Bipolar Disorder. ACS Chem Neurosci 2019; 10:1214-1221. [PMID: 30644730 DOI: 10.1021/acschemneuro.8b00629] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Bipolar disorder is a complex neuropsychiatric disorder, characterized by intermittent episodes of mania and depression. Recent studies have indicated argyrophilic grains, composed of hyperphosphorylated tau, are observable in postmortem brains of bipolar patients. It remains uncertain how tau hyperphosphorylation results in neurodegeneration upon the disease. Recent studies have demonstrated that phosphorylated tau at Thr231 exists in two distinct cis and trans conformations, in which cis pT231-tau is highly neurotoxic and acts as an early driver of tauopathy in several neurodegenerative diseases. We herein employed an in vitro model, which resembles some aspects of bipolar disorder, to study the cis p-tau mediatory role. We established GSK3β overexpressing SH-SY5Y cells and examined cell viability, cis p-tau formation, and lithium effects by immunofluorescence and flow cytometry. We found an increase in cis p-tau levels as well as viability decrease in the cell model. Furthermore, we discovered that lithium treatment inhibits cis p-tau formation, resulting in diminished cell death. We also examined BD and healthy human brain samples and detected cis p-tau in the patients' brains. Our results show that tauopathy, observed in bipolar disorder, is being mediated through cis p-tau and that a conformer could be the cause of neurodegeneration upon the disease. Our findings would suggest novel therapeutic target to fight the devastating disorder.
Collapse
Affiliation(s)
- Roghayeh Naserkhaki
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Selva Zamanzadeh
- Department of Molecular Biology, Islamic Azad University, Tehran Medical Sciences Branch, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Seyed Massood Nabavi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Pakdaman
- Department of Neurology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahba Shahbazi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Massood Vosough
- Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Gholam Ghaedi
- Mostafa Khomeini Hospital, School of Medicine, Shahed University, Tehran, Iran
| | | | - Davood Mirtorabi
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Mohamad Hedayatshodeh
- Department of Emergency Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Ehsani
- Department of Biology, Roudehen Branch, Islamic Azad University, Roudehen, Iran
| | - Mojtaba Falahati
- Department of Nanotechnology, Faculty of Advance Science and Technology, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Javad Hajipour
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Koorosh Shahpasand
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
41
|
Mecocci P, Baroni M, Senin U, Boccardi V. Brain Aging and Late-Onset Alzheimer’s Disease: A Matter of Increased Amyloid or Reduced Energy? J Alzheimers Dis 2018; 64:S397-S404. [DOI: 10.3233/jad-179903] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Patrizia Mecocci
- Department of Medicine, Institute of Gerontology and Geriatrics, University of Perugia, Italy
| | - Marta Baroni
- Department of Medicine, Institute of Gerontology and Geriatrics, University of Perugia, Italy
| | - Umberto Senin
- Department of Medicine, Institute of Gerontology and Geriatrics, University of Perugia, Italy
| | - Virginia Boccardi
- Department of Medicine, Institute of Gerontology and Geriatrics, University of Perugia, Italy
| |
Collapse
|
42
|
Therapeutic strategies for targeting neurodegenerative protein misfolding disorders. Curr Opin Chem Biol 2018; 44:66-74. [PMID: 29902695 DOI: 10.1016/j.cbpa.2018.05.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 05/25/2018] [Indexed: 12/14/2022]
Abstract
Neurodegenerative diseases can arise from a multitude of different pathological drivers, however protein misfolding appears to be a common molecular feature central to several disorders. Protein folding, and attainment of correct secondary and tertiary structure, is essential for proper protein function. Protein misfolding gives rise to structural perturbations that can result in loss of protein function or a gain of toxic function, such as through aggregation, either of which can initiate and propagate biological responses that are deleterious to cells. Several neurodegenerative diseases, such as Alzheimer's disease, amyotrophic lateral sclerosis, Huntington's disease and Parkinson's disease, each have identified molecular components in which protein misfolding perturbs cellular systems that ultimately lead to cell death, and this predominately occurs in neurons. Current efforts focused on developing therapies for protein misfolding disorders have employed diverse strategies; inhibiting the production of disease-relevant proteins prone to misfolding, inhibiting the aggregation of misfolded proteins, removing and preventing spread of aggregated misfolded proteins and manipulating cellular systems to mitigate the toxic effects of misfolded proteins. Each of these strategies has yielded therapeutic agents that have transitioned from preclinical proof of concept studies into human clinical testing. These approaches and therapies are described herein.
Collapse
|
43
|
Ferrero H, Larrayoz IM, Gil-Bea FJ, Martínez A, Ramírez MJ. Adrenomedullin, a Novel Target for Neurodegenerative Diseases. Mol Neurobiol 2018; 55:8799-8814. [PMID: 29600350 DOI: 10.1007/s12035-018-1031-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 03/22/2018] [Indexed: 01/18/2023]
Abstract
Neurodegenerative diseases represent a heterogeneous group of disorders whose common characteristic is the progressive degeneration of neuronal structure and function. Although much knowledge has been accumulated on the pathophysiology of neurodegenerative diseases over the years, more efforts are needed to understand the processes that underlie these diseases and hence to propose new treatments. Adrenomedullin (AM) is a multifunctional peptide involved in vasodilation, hormone secretion, antimicrobial defense, cellular growth, and angiogenesis. In neurons, AM and related peptides are associated with some structural and functional cytoskeletal proteins that interfere with microtubule dynamics. Furthermore, AM may intervene in neuronal dysfunction through other mechanisms such as immune and inflammatory response, apoptosis, or calcium dyshomeostasis. Alterations in AM expression have been described in neurodegenerative processes such as Alzheimer's disease or vascular dementia. This review addresses the current state of knowledge on AM and its possible implication in neurodegenerative diseases.
Collapse
Affiliation(s)
- Hilda Ferrero
- Department of Pharmacology and Toxicology, and IdiSNA, Navarra Institute for Health Research, University of Navarra, Pamplona, Spain
| | - Ignacio M Larrayoz
- Biomarkers and Molecular Signaling, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - Francisco J Gil-Bea
- Department of Pharmacology and Toxicology, and IdiSNA, Navarra Institute for Health Research, University of Navarra, Pamplona, Spain
- Neuroscience Area, Biodonostia Health Research Institute, CIBERNED, San Sebastian, Spain
| | - Alfredo Martínez
- Oncology Area, Center for Biomedical Research of La Rioja (CIBIR), Logroño, Spain
| | - María J Ramírez
- Department of Pharmacology and Toxicology, and IdiSNA, Navarra Institute for Health Research, University of Navarra, Pamplona, Spain.
| |
Collapse
|
44
|
Diabetes mellitus and Alzheimer’s disease: GSK-3β as a potential link. Behav Brain Res 2018; 339:57-65. [DOI: 10.1016/j.bbr.2017.11.015] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 10/08/2017] [Accepted: 11/13/2017] [Indexed: 11/19/2022]
|
45
|
Jan AT, Azam M, Rahman S, Almigeiti AMS, Choi DH, Lee EJ, Haq QMR, Choi I. Perspective Insights into Disease Progression, Diagnostics, and Therapeutic Approaches in Alzheimer's Disease: A Judicious Update. Front Aging Neurosci 2017; 9:356. [PMID: 29163138 PMCID: PMC5671974 DOI: 10.3389/fnagi.2017.00356] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 10/18/2017] [Indexed: 01/22/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the progressive accumulation of β-amyloid fibrils and abnormal tau proteins in and outside of neurons. Representing a common form of dementia, aggravation of AD with age increases the morbidity rate among the elderly. Although, mutations in the ApoE4 act as potent risk factors for sporadic AD, familial AD arises through malfunctioning of APP, PSEN-1, and−2 genes. AD progresses through accumulation of amyloid plaques (Aβ) and neurofibrillary tangles (NFTs) in brain, which interfere with neuronal communication. Cellular stress that arises through mitochondrial dysfunction, endoplasmic reticulum malfunction, and autophagy contributes significantly to the pathogenesis of AD. With high accuracy in disease diagnostics, Aβ deposition and phosphorylated tau (p-tau) are useful core biomarkers in the cerebrospinal fluid (CSF) of AD patients. Although five drugs are approved for treatment in AD, their failures in achieving complete disease cure has shifted studies toward a series of molecules capable of acting against Aβ and p-tau. Failure of biologics or compounds to cross the blood-brain barrier (BBB) in most cases advocates development of an efficient drug delivery system. Though liposomes and polymeric nanoparticles are widely adopted for drug delivery modules, their use in delivering drugs across the BBB has been overtaken by exosomes, owing to their promising results in reducing disease progression.
Collapse
Affiliation(s)
- Arif Tasleem Jan
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Mudsser Azam
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Safikur Rahman
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Angham M S Almigeiti
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Duk Hwan Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | | | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| |
Collapse
|
46
|
Insight into the mechanism of action and selectivity of caspase-3 reversible inhibitors through in silico studies. J Mol Struct 2017. [DOI: 10.1016/j.molstruc.2017.06.118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
47
|
Nizynski B, Dzwolak W, Nieznanski K. Amyloidogenesis of Tau protein. Protein Sci 2017; 26:2126-2150. [PMID: 28833749 DOI: 10.1002/pro.3275] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/16/2017] [Accepted: 08/16/2017] [Indexed: 11/08/2022]
Abstract
The role of microtubule-associated protein Tau in neurodegeneration has been extensively investigated since the discovery of Tau amyloid aggregates in the brains of patients with Alzheimer's disease (AD). The process of formation of amyloid fibrils is known as amyloidogenesis and attracts much attention as a potential target in the prevention and treatment of neurodegenerative conditions linked to protein aggregation. Cerebral deposition of amyloid aggregates of Tau is observed not only in AD but also in numerous other tauopathies and prion diseases. Amyloidogenesis of intrinsically unstructured monomers of Tau can be triggered by mutations in the Tau gene, post-translational modifications, or interactions with polyanionic molecules and aggregation-prone proteins/peptides. The self-assembly of amyloid fibrils of Tau shares a number of characteristic features with amyloidogenesis of other proteins involved in neurodegenerative diseases. For example, in vitro experiments have demonstrated that the nucleation phase, which is the rate-limiting stage of Tau amyloidogenesis, is shortened in the presence of fragmented preformed Tau fibrils acting as aggregation templates ("seeds"). Accordingly, Tau aggregates released by tauopathy-affected neurons can spread the neurodegenerative process in the brain through a prion-like mechanism, originally described for the pathogenic form of prion protein. Moreover, Tau has been shown to form amyloid strains-structurally diverse self-propagating aggregates of potentially various pathological effects, resembling in this respect prion strains. Here, we review the current literature on Tau aggregation and discuss mechanisms of propagation of Tau amyloid in the light of the prion-like paradigm.
Collapse
Affiliation(s)
- Bartosz Nizynski
- College of Inter-Faculty Individual Studies in Mathematics and Natural Sciences, University of Warsaw, 2C Banacha Str, Warsaw, 02-097, Poland.,Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, 1 Pasteur Str, Warsaw, 02-093, Poland
| | - Wojciech Dzwolak
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, 1 Pasteur Str, Warsaw, 02-093, Poland
| | - Krzysztof Nieznanski
- Department of Biochemistry, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Str, Warsaw, 02-093, Poland
| |
Collapse
|
48
|
Ferrero K, Silver M, Cocchetto A, Masliah E, Langford D. CNS findings in chronic fatigue syndrome and a neuropathological case report. J Investig Med 2017; 65:974-983. [PMID: 28386034 DOI: 10.1136/jim-2016-000390] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2017] [Indexed: 11/04/2022]
Abstract
Chronic fatigue syndrome (CFS) is characterized as a persistent, debilitating complex disorder of unknown etiology, whereby patients suffer from extreme fatigue, which often presents with symptoms that include chronic pain, depression, weakness, mood disturbances, and neuropsychological impairment. In this mini review and case report, we address central nervous system (CNS) involvement of CFS and present neuropathological autopsy findings from a patient who died with a prior diagnosis of CFS. Among the most remarkable pathological features of the case are focal areas of white matter loss, neurite beading, and neuritic pathology of axons in the white matter with axonal spheroids. Atypical neurons displaying aberrant sprouting processes in response to injury are observed throughout cortical gray and white matter. Abundant amyloid deposits identical to AD plaques with accompanying intracellular granular structures are observed as well. Neurofibrillary tangles are also present in the white matter of the frontal cortex, thalamus and basal ganglia. Taken together, these neuropathological findings warrant further studies into CNS disease associated with CFS.
Collapse
Affiliation(s)
- Kimberly Ferrero
- Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Mitchell Silver
- Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Alan Cocchetto
- State University of New York at Alfred, Engineering Technologies, Alfred, New York, USA
| | - Eliezer Masliah
- University of California San Diego, La Jolla, California, USA
| | - Dianne Langford
- Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
49
|
Gender-Specific Expression of Ubiquitin-Specific Peptidase 9 Modulates Tau Expression and Phosphorylation: Possible Implications for Tauopathies. Mol Neurobiol 2016; 54:7979-7993. [PMID: 27878758 PMCID: PMC5684262 DOI: 10.1007/s12035-016-0299-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 11/14/2016] [Indexed: 01/05/2023]
Abstract
Public transcriptomic studies have shown that several genes display pronounced gender differences in their expression in the human brain, which may influence the manifestations and risk for neuronal disorders. Here, we apply a transcriptome-wide analysis to discover genes with gender-specific expression and significant alterations in public postmortem brain tissue from Alzheimer’s disease (AD) patients compared to controls. We identify the sex-linked ubiquitin-specific peptidase 9 (USP9) as an outstanding candidate gene with highly significant expression differences between the genders and male-specific underexpression in AD. Since previous studies have shown that USP9 can modulate the phosphorylation of the AD-associated protein MAPT, we investigate functional associations between USP9 and MAPT in further detail. After observing a high positive correlation between the expression of USP9 and MAPT in the public transcriptomics data, we show that USP9 knockdown results in significantly decreased MAPT expression in a DU145 cell culture model and a concentration-dependent decrease for the MAPT orthologs mapta and maptb in a zebrafish model. From the analysis of microarray and qRT-PCR experiments for the knockdown in DU145 cells and prior knowledge from the literature, we derive a data-congruent model for a USP9-dependent regulatory mechanism modulating MAPT expression via BACH1 and SMAD4. Overall, the analyses suggest USP9 may contribute to molecular gender differences observed in tauopathies and provide a new target for intervention strategies to modulate MAPT expression.
Collapse
|
50
|
Sawatzky E, Wehle S, Kling B, Wendrich J, Bringmann G, Sotriffer CA, Heilmann J, Decker M. Discovery of Highly Selective and Nanomolar Carbamate-Based Butyrylcholinesterase Inhibitors by Rational Investigation into Their Inhibition Mode. J Med Chem 2016; 59:2067-82. [DOI: 10.1021/acs.jmedchem.5b01674] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Edgar Sawatzky
- Pharmazeutische
und Medizinische Chemie, Institut für Pharmazie und Lebensmittelchemie, Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Sarah Wehle
- Pharmazeutische
und Medizinische Chemie, Institut für Pharmazie und Lebensmittelchemie, Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Beata Kling
- Lehrstuhl
für Pharmazeutische Biologie, Institut für Pharmazie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Jan Wendrich
- Lehrstuhl
für Organische Chemie I, Institut für Organische Chemie, Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Gerhard Bringmann
- Lehrstuhl
für Organische Chemie I, Institut für Organische Chemie, Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Christoph A. Sotriffer
- Pharmazeutische
und Medizinische Chemie, Institut für Pharmazie und Lebensmittelchemie, Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Jörg Heilmann
- Lehrstuhl
für Pharmazeutische Biologie, Institut für Pharmazie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Michael Decker
- Pharmazeutische
und Medizinische Chemie, Institut für Pharmazie und Lebensmittelchemie, Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| |
Collapse
|