151
|
Dinius CJ, Pocknell CE, Caffrey MP, Roche RAP. Cognitive interventions for memory and psychological well-being in aging and dementias. Front Psychol 2023; 14:1070012. [PMID: 36818134 PMCID: PMC9932670 DOI: 10.3389/fpsyg.2023.1070012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/17/2023] [Indexed: 02/05/2023] Open
Abstract
The human lifespan has expanded drastically in the last few centuries, due to improvements in sanitation, medicine, and nutrition, but with this increase in longevity comes higher rates of cognitive pathology such as mild cognitive impairment (MCI) and dementia; the latter is estimated to reach more than 75 million people by 2030. Pathology risk is related to measures of executive function, lifestyle factors (e.g., education, occupation, and leisure activities), and cognitive reserve. One way of building cognitive reserve may be to structure the environment to encourage lifelong engagement and learning, and since a pharmacological "cure" for dementia remains elusive, non-pharmacological approaches such as physical activity, social engagement, and cognitive stimulation are becoming increasingly essential to preserving and protecting brain health. Here, we describe our recent research into Reminiscence Therapy (RT) to promote cognitive and psychological function in old age and early dementia. We review the Recall Initiative, which involved brain imaging and behavioral indices of memory pre- and post-RT. We also report results from a pilot study-AIM WARM-in which RT was combined with physical activity, specifically walking, for early-stage dementia. Finally, we outline our future directions for tailored reminiscence interventions in combination with other activities (e.g., yoga and meditation) for different groups, namely early Alzheimer's disease, Semantic Dementia, and older individuals in the prison system.
Collapse
|
152
|
Li Q, Xiao M, Li N, Cai W, Zhao C, Liu B, Zeng F. Application of
Caenorhabditis elegans
in the evaluation of food nutrition: A review. EFOOD 2023. [DOI: 10.1002/efd2.68] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
Affiliation(s)
- Quancen Li
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Meifang Xiao
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Na Li
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Wenwen Cai
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Chao Zhao
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
- College of Marine Sciences Fujian Agriculture and Forestry University Fuzhou China
- Engineering Research Center of Fujian Subtropical Fruit and Vegetable Processing Fujian Agriculture and Forestry University Fuzhou China
| | - Bin Liu
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
- Engineering Research Center of Fujian Subtropical Fruit and Vegetable Processing Fujian Agriculture and Forestry University Fuzhou China
- National Engineering Research Center of JUNCAO Technology Fujian Agriculture and Forestry University Fuzhou China
| | - Feng Zeng
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
- Engineering Research Center of Fujian Subtropical Fruit and Vegetable Processing Fujian Agriculture and Forestry University Fuzhou China
| |
Collapse
|
153
|
Yan M, Wang L, Wu Y, Wang L, Lu Y. Three-dimensional highly porous hydrogel scaffold for neural circuit dissection and modulation. Acta Biomater 2023; 157:252-262. [PMID: 36521677 DOI: 10.1016/j.actbio.2022.12.011] [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: 08/11/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Biomimetic brain structures and artificial neural networks have provided a simplified strategy for quantitatively investigating the complex structural and functional characteristics of highly interconnected neural networks. To achieve this, three-dimensional (3D) cell culture approaches have attracted much attention, which can mimic cell-cell interactions at the organism level and help better understand the function of specific neurons and neuronal networks than traditional two-dimensional cell culture methods. However, 3D scaffolds similar to the natural extracellular matrix to support the culturing, recording, and manipulation of neurons have long been an unresolved challenge. To resolve this, 3D hydrogel scaffolds can be fabricated via an innovative thermal treatment followed by an esterification process. A highly porous microstructure was formed within the bulk hydrogel scaffold, which showed a high porosity of 91% and a low Young's modulus of 6.11 kPa. Due to the merits of the fabricated hydrogel scaffolds, we constructed 3D neural networks and detected spontaneous action potentials in vitro. We successfully induced seizure-like waveforms in 3D cultured neurons and suppressed hyperactivated discharges by selectively activating γ-aminobutyric acid-ergic (GABAergic) interneurons. These results prove the advantages of our hydrogel scaffolds and demonstrate their application potential in the accurate dissection of neural circuits, which may help develop effective treatments for various neurological disorders. STATEMENT OF SIGNIFICANCE: While 3D cell culture approaches have attracted much attention and offer more advantages than two-dimensional cell culture methods, 3D scaffolds similar to the natural extracellular matrix to support the culturing, recording, and manipulation of neurons have long been an unresolved challenge. Herein, we developed a simplified and low-cost strategy for fabricating highly porous and cytocompatible hydrogel scaffolds for the construction of three-dimensional (3D) neural networks in vitro. The cultured 3D neural networks can mimic the in vivo connection among different neuron subgroups and help accurately dissect and manipulate the structure and function of specific neural circuits.
Collapse
Affiliation(s)
- Mengying Yan
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Lulu Wang
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Yiyong Wu
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Liping Wang
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China.
| | - Yi Lu
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China.
| |
Collapse
|
154
|
Alonso Torrens A, Mitchell CA, Pourshahidi LK, Murphy BÓ, Allwood W, Rizzetto L, Scholz M, Tuohy K, Pereira-Caro G, Moreno-Rojas JM, McDougall G, Gill CIR. Long-term supplementation with anthocyanin-rich or -poor Rubus idaeus berries does not influence microvascular architecture nor cognitive outcome in the APP/PS-1 mouse model of Alzheimer's disease. Int J Food Sci Nutr 2023; 74:33-50. [PMID: 36450698 DOI: 10.1080/09637486.2022.2141209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Disruption of microvascular architecture is a common pathogenic mechanism in the progression of Alzheimer's disease (AD). Given the anti-angiogenic activity of berry (poly)phenols, we investigated whether long-term feeding of Rubus idaeus (raspberries) could ameliorate cerebral microvascular pathology and improve cognition in the APP/PS-1 mouse model of AD. Male C57Bl/6J mice (50 wild type, 50 APP/PS-1) aged 4-months were fed for 24-weeks, with a normal diet enriched with either 100 mg/day glucose (control diet) or supplemented with glucose and freeze-dried anthocyanin-rich (red) or -poor (yellow) raspberries (100 mg/day) and assessed/sampled post intervention. Cerebral microvascular architecture of wild-type mice was characterised by regularly spaced capillaries with uniform diameters, unlike APP/PS-1 transgenic mice which showed dysregulated microvascular architecture. Long-term feeding of raspberries demonstrated limited modulation of microbiota and no substantive effect on microvascular architecture or cognition in either mice model although changes were evident in endogenous cerebral and plasmatic metabolites.
Collapse
Affiliation(s)
- Aaron Alonso Torrens
- Nutrition Innovation Centre for Food and Health (NICHE), Centre for Molecular Biosciences, University of Ulster, Coleraine, Northern Ireland, UK
| | - Christopher A Mitchell
- Nutrition Innovation Centre for Food and Health (NICHE), Centre for Molecular Biosciences, University of Ulster, Coleraine, Northern Ireland, UK
| | - L Kirsty Pourshahidi
- Nutrition Innovation Centre for Food and Health (NICHE), Centre for Molecular Biosciences, University of Ulster, Coleraine, Northern Ireland, UK
| | - Brian Óg Murphy
- Nutrition Innovation Centre for Food and Health (NICHE), Centre for Molecular Biosciences, University of Ulster, Coleraine, Northern Ireland, UK
| | - William Allwood
- Plant Biochemistry and Food Quality Group, Environmental and Biochemical Sciences, The James Hutton Institute, Invergowrie, Dundee, Scotland
| | - Lisa Rizzetto
- Nutrition and Nutrigenomics Unit, Research and Innovation Centre, San Michele all'Adige, Trentino, Italy
| | - Matthias Scholz
- Nutrition and Nutrigenomics Unit, Research and Innovation Centre, San Michele all'Adige, Trentino, Italy
| | - Kieran Tuohy
- Nutrition and Nutrigenomics Unit, Research and Innovation Centre, San Michele all'Adige, Trentino, Italy
| | - Gema Pereira-Caro
- Department of Food Science and Health, Andalusian Institute of Agricultural and Fisheries Research and Training (IFAPA), Alameda del Obispo, Córdoba, Spain.,Foods for Health Group, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
| | - José Manuel Moreno-Rojas
- Department of Food Science and Health, Andalusian Institute of Agricultural and Fisheries Research and Training (IFAPA), Alameda del Obispo, Córdoba, Spain.,Foods for Health Group, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
| | - Gordon McDougall
- Plant Biochemistry and Food Quality Group, Environmental and Biochemical Sciences, The James Hutton Institute, Invergowrie, Dundee, Scotland
| | - Chris I R Gill
- Nutrition Innovation Centre for Food and Health (NICHE), Centre for Molecular Biosciences, University of Ulster, Coleraine, Northern Ireland, UK
| |
Collapse
|
155
|
Kilic B, Bardakkaya M, Ilıkcı Sagkan R, Aksakal F, Shakila S, Dogruer DS. New thiourea and benzamide derivatives of 2-aminothiazole as multi-target agents against Alzheimer's disease: Design, synthesis, and biological evaluation. Bioorg Chem 2023; 131:106322. [PMID: 36565675 DOI: 10.1016/j.bioorg.2022.106322] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/30/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022]
Abstract
In this study, two series of compounds were designed and synthesized, bearing thiourea and benzamide derivatives at position 2 of 4-subtituted-2-aminothiazole, respectively. Then, the inhibition potency of all final compounds for cholinesterase enzymes were evaluated. Among the thiourea derivatives, 3c (IC50 = 0.33 μM) was identified as the most potent and selective butyrylcholinesterase inhibitor. Additionally, benzamide derivative 10e (AChE IC50 = 1.47 and BChE IC50 = 11.40 μM) was found as a dual cholinesterase inhibitor. The type of inhibition for both compounds was determined by kinetic studies and the results showed that the compounds were mixed type inhibitors. Moreover, all title compounds were investigated in terms of their antioxidant (DPHH, ORAC) and metal chelator activities. In addition, the neuroprotective effects of selected compounds (3c, 3e, 6c, 6e and 10e) against H2O2-induced damage in the PC12 cell line were tested. The experimental findings demonstrated that thiourea-derived 6e (40.4 %) and benzamide-derived 10e (37.8 %) have a neuroprotective effect of about half as ferulic acid at 10 μM. Subsequently, the cytotoxicity of selected compounds was examined by the MTT assay, and the compounds were found not to have cytotoxic effect on the PC12 cell line in 24 h. Additionally, compounds 6e and 10e were also found to be more effective in inhibiting the release of IL-1β, IL-6, TNF-α and NO compared to other selected compounds in this study.
Collapse
Affiliation(s)
- Burcu Kilic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Ankara, Turkiye
| | - Merve Bardakkaya
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Ankara, Turkiye; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Biruni University, İstanbul, Turkiye
| | - Rahsan Ilıkcı Sagkan
- Department of Medical Biology, Faculty of Medicine, Uşak University, Uşak, Turkiye
| | - Fatma Aksakal
- Department of Chemistry, Hacettepe University, Ankara, Turkiye; Department of Analytical Chemistry, Faculty of Pharmacy, Kocaeli Health and Technology University, Kocaeli, Turkiye
| | - Shakila Shakila
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Ankara, Turkiye
| | - Deniz S Dogruer
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Ankara, Turkiye.
| |
Collapse
|
156
|
Sridharan B, Lim HG. Exosomes and ultrasound: The future of theranostic applications. Mater Today Bio 2023; 19:100556. [PMID: 36756211 PMCID: PMC9900624 DOI: 10.1016/j.mtbio.2023.100556] [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: 11/17/2022] [Revised: 01/17/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Biomaterials and pertaining formulations have been very successful in various diagnostic and therapeutic applications because of its ability to overcome pharmacological limitations. Some of them have gained significant focus in the recent decade for their theranostic properties. Exosomes can be grouped as biomaterials, since they consist of various biological micro/macromolecules and possess all the properties of a stable biomaterial with size in nano range. Significant research has gone into isolation and exploitation of exosomes as potential theranostic agent. However, the limitations in terms of yield, efficacy, and target specificity are continuously being addressed. On the other hand, several nano/microformulations are responsive to physical or chemical alterations and were successfully stimulated by tweaking the physical characteristics of the surrounding environment they are in. Some of them are termed as photodynamic, sonodynamic or thermodynamic therapeutic systems. In this regard, ultrasound and acoustic systems were extensively studied for its ability towards altering the properties of the systems to which they were applied on. In this review, we have detailed about the diagnostic and therapeutic applications of exosomes and ultrasound separately, consisting of their conventional applications, drawbacks, and developments for addressing the challenges. The information were categorized into various sections that provide complete overview of the isolation strategies and theranostic applications of exosomes in various diseases. Then the ultrasound-based disease diagnosis and therapy were elaborated, with special interest towards the use of ultrasound in enhancing the efficacy of nanomedicines and nanodrug delivery systems, Finally, we discussed about the ability of ultrasound in enhancing the diagnostic and therapeutic properties of exosomes, which could be the future of theranostics.
Collapse
Affiliation(s)
| | - Hae Gyun Lim
- Corresponding author. Biomedical Ultrasound Lab, Department of Biomedical Engineering, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
157
|
Liu P, Cheng M, Guo J, Cao D, Luo J, Wan Y, Fang Y, Jin Y, Xie SS, Liu J. Dual functional antioxidant and butyrylcholinesterase inhibitors for the treatment of Alzheimer's disease: Design, synthesis and evaluation of novel melatonin-alkylbenzylamine hybrids. Bioorg Med Chem 2023; 78:117146. [PMID: 36580744 DOI: 10.1016/j.bmc.2022.117146] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022]
Abstract
Here, we have designed and synthesized a series of melatonin-alkylbenzylamine hybrids as multitarget agents for the treatment of Alzheimer's disease (AD). Most of them exhibited a potent multifunctional profile involving cholinesterase inhibition and antioxidant effects. Among these compounds, compound 5 was most the potent antioxidant (ORAC = 5.13) and also an excellent selective inhibitor of BuChE (huBuChE IC50 = 1.20 μM, huAChE IC50 = 177.49 μM, SI = 147.91). Moreover, kinetic study indicated compound 5 was a mixed-type inhibitor for huBuChE. Furthermore, it could induce expression of the Nrf2 as well as its downstream markers at the protein level in cells. More importantly, compound 5 display no acute toxicity in mice at doses up to 2500 mg/kg. And we found compound 5 could improve memory function of scopolamine-induced amnesia mice. These results highlighted compound 5 as a possible hit molecule for further investigation of new anti-AD drugs.
Collapse
Affiliation(s)
- Peng Liu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330006, PR China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Maojun Cheng
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330006, PR China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Jie Guo
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Duanyuan Cao
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Jinchong Luo
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Yang Wan
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Yuanying Fang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Yi Jin
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Sai-Sai Xie
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, PR China.
| | - Jing Liu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330006, PR China.
| |
Collapse
|
158
|
Meng S, Chen H, Deng C, Meng Z. Catalpol Mitigates Alzheimer's Disease Progression by Promoting the Expression of Neural Stem Cell Exosomes Released miR-138-5p. Neurotox Res 2023; 41:41-56. [PMID: 36595161 PMCID: PMC9944361 DOI: 10.1007/s12640-022-00626-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/11/2022] [Accepted: 12/16/2022] [Indexed: 01/04/2023]
Abstract
Alzheimer's disease (Alzheimer's disease, AD) is a neurodegenerative disease characterized by senile plaque deposition and neurofibrillary tangles. The pathogenesis of AD is complicated and the drugs used to treat AD are single-targeted drugs, which can only improve or alleviate the symptoms of patients, but cannot delay or prevent the progress of the disease. Because of its ability to act on multiple targets, multiple systems, multiple links, and multiple pathways, Chinese herbal compound prescriptions have shown unique advantages in the research and treatment of AD. Our previous study has demonstrated the protect role of the Chinese medicine Rehmannia in AD. However, the underlying mechanism remains unclear. In the present study, both in vitro and vivo experiments were employed, and we found Catalpol (Ca), the main extract of Rehmannia, could mitigate AD progression both in vitro and in vivo by promoting miR-138-5p level in neural stem cell secreted exosomes.
Collapse
Affiliation(s)
- Shengxi Meng
- Department of Traditional Chinese Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Xuhui District, No.600 Yi Shan Road, Shanghai, 200233, China.
| | - Huize Chen
- Department of Traditional Chinese Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Xuhui District, No.600 Yi Shan Road, Shanghai, 200233, China
| | - Chunjun Deng
- Department of Traditional Chinese Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Xuhui District, No.600 Yi Shan Road, Shanghai, 200233, China
| | - Zeyu Meng
- Second Clinical Medicine College, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| |
Collapse
|
159
|
Sousa JA, Bernardes C, Bernardo-Castro S, Lino M, Albino I, Ferreira L, Brás J, Guerreiro R, Tábuas-Pereira M, Baldeiras I, Santana I, Sargento-Freitas J. Reconsidering the role of blood-brain barrier in Alzheimer's disease: From delivery to target. Front Aging Neurosci 2023; 15:1102809. [PMID: 36875694 PMCID: PMC9978015 DOI: 10.3389/fnagi.2023.1102809] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
The existence of a selective blood-brain barrier (BBB) and neurovascular coupling are two unique central nervous system vasculature features that result in an intimate relationship between neurons, glia, and blood vessels. This leads to a significant pathophysiological overlap between neurodegenerative and cerebrovascular diseases. Alzheimer's disease (AD) is the most prevalent neurodegenerative disease whose pathogenesis is still to be unveiled but has mostly been explored under the light of the amyloid-cascade hypothesis. Either as a trigger, bystander, or consequence of neurodegeneration, vascular dysfunction is an early component of the pathological conundrum of AD. The anatomical and functional substrate of this neurovascular degeneration is the BBB, a dynamic and semi-permeable interface between blood and the central nervous system that has consistently been shown to be defective. Several molecular and genetic changes have been demonstrated to mediate vascular dysfunction and BBB disruption in AD. The isoform ε4 of Apolipoprotein E is at the same time the strongest genetic risk factor for AD and a known promoter of BBB dysfunction. Low-density lipoprotein receptor-related protein 1 (LRP-1), P-glycoprotein, and receptor for advanced glycation end products (RAGE) are examples of BBB transporters implicated in its pathogenesis due to their role in the trafficking of amyloid-β. This disease is currently devoid of strategies that change the natural course of this burdening illness. This unsuccess may partly be explained by our misunderstanding of the disease pathogenesis and our inability to develop drugs that are effectively delivered to the brain. BBB may represent a therapeutic opportunity as a target itself or as a therapeutic vehicle. In this review, we aim to explore the role of BBB in the pathogenesis of AD including the genetic background and detail how it can be targeted in future therapeutic research.
Collapse
Affiliation(s)
- João André Sousa
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Catarina Bernardes
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Sara Bernardo-Castro
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Miguel Lino
- Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Inês Albino
- Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Lino Ferreira
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - José Brás
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| | - Rita Guerreiro
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| | - Miguel Tábuas-Pereira
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Inês Baldeiras
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - Isabel Santana
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - João Sargento-Freitas
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
160
|
Chen X, Jiang S, Wang R, Bao X, Li Y. Neural Stem Cells in the Treatment of Alzheimer's Disease: Current Status, Challenges, and Future Prospects. J Alzheimers Dis 2023; 94:S173-S186. [PMID: 36336934 PMCID: PMC10473082 DOI: 10.3233/jad-220721] [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] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
Alzheimer's disease (AD), a progressive dementia, is one of the world's most dangerous and debilitating diseases. Clinical trial results of amyloid-β (Aβ) and tau regulators based on the pretext of straightforward amyloid and tau immunotherapy were disappointing. There are currently no effective strategies for slowing the progression of AD. Further understanding of the mechanisms underlying AD and the development of novel therapeutic options are critical. Neurogenesis is impaired in AD, which contributes to memory deficits. Transplanted neural stem cells (NSCs) can regenerate degraded cholinergic neurons, and new neurons derived from NSCs can form synaptic connections with neighboring neurons. In theory, employing NSCs to replace and restore damaged cholinergic neurons and brain connections may offer new treatment options for AD. However there remain barriers to surmount before NSC-based therapy can be used clinically. The objective of this article is to describe recent advances in the treatment of AD models and clinical trials involving NSCs. In addition, we discuss the challenges and prospects associated with cell transplant therapy for AD.
Collapse
Affiliation(s)
- Xiaokun Chen
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Shenzhong Jiang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xinjie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yongning Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
161
|
Wang Y, Hu H, Liu X, Guo X. Hypoglycemic medicines in the treatment of Alzheimer's disease: Pathophysiological links between AD and glucose metabolism. Front Pharmacol 2023; 14:1138499. [PMID: 36909158 PMCID: PMC9995522 DOI: 10.3389/fphar.2023.1138499] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Alzheimer's Disease (AD) is a global chronic disease in adults with beta-amyloid (Aβ) deposits and hyperphosphorylated tau protein as the pathologic characteristics. Although the exact etiology of AD is still not fully elucidated, aberrant metabolism including insulin signaling and mitochondria dysfunction plays an important role in the development of AD. Binding to insulin receptor substrates, insulin can transport through the blood-brain barrier (BBB), thus mediating insulin signaling pathways to regulate physiological functions. Impaired insulin signaling pathways, including PI3K/Akt/GSK3β and MAPK pathways, could cause damage to the brain in the pathogenesis of AD. Mitochondrial dysfunction and overexpression of TXNIP could also be causative links between AD and DM. Some antidiabetic medicines may have benefits in the treatment of AD. Metformin can be beneficial for cognition improvement in AD patients, although results from clinical trials were inconsistent. Exendin-4 may affect AD in animal models but there is a lack of clinical trials. Liraglutide and dulaglutide could also benefit AD patients in adequate clinical studies but not semaglutide. Dipeptidyl peptidase IV inhibitors (DPP4is) such as saxagliptin, vildagliptin, linagliptin, and sitagliptin could boost cognitive function in animal models. And SGLT2 inhibitors such as empagliflozin and dapagliflozin were also considerably protective against new-onset dementia in T2DM patients. Insulin therapy is a promising therapy but some studies indicated that it may increase the risk of AD. Herbal medicines are helpful for cognitive function and neuroprotection in the brain. For example, polyphenols, alkaloids, glycosides, and flavonoids have protective benefits in cognition function and glucose metabolism. Focusing on glucose metabolism, we summarized the pharmacological mechanism of hypoglycemic drugs and herbal medicines. New treatment approaches including antidiabetic synthesized drugs and herbal medicines would be provided to patients with AD. More clinical trials are needed to produce definite evidence for the effectiveness of hypoglycemic medications.
Collapse
Affiliation(s)
- Yixuan Wang
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Hao Hu
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Xinyu Liu
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Xiangyu Guo
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
162
|
Akhtar A, Gupta SM, Dwivedi S, Kumar D, Shaikh MF, Negi A. Preclinical Models for Alzheimer's Disease: Past, Present, and Future Approaches. ACS OMEGA 2022; 7:47504-47517. [PMID: 36591205 PMCID: PMC9798399 DOI: 10.1021/acsomega.2c05609] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/22/2022] [Indexed: 05/13/2023]
Abstract
A robust preclinical disease model is a primary requirement to understand the underlying mechanisms, signaling pathways, and drug screening for human diseases. Although various preclinical models are available for several diseases, clinical models for Alzheimer's disease (AD) remain underdeveloped and inaccurate. The pathophysiology of AD mainly includes the presence of amyloid plaques and neurofibrillary tangles (NFT). Furthermore, neuroinflammation and free radical generation also contribute to AD. Currently, there is a wide gap in scientific approaches to preventing AD progression. Most of the available drugs are limited to symptomatic relief and improve deteriorating cognitive functions. To mimic the pathogenesis of human AD, animal models like 3XTg-AD and 5XFAD are the primarily used mice models in AD therapeutics. Animal models for AD include intracerebroventricular-streptozotocin (ICV-STZ), amyloid beta-induced, colchicine-induced, etc., focusing on parameters such as cognitive decline and dementia. Unfortunately, the translational rate of the potential drug candidates in clinical trials is poor due to limitations in imitating human AD pathology in animal models. Therefore, the available preclinical models possess a gap in AD modeling. This paper presents an outline that critically assesses the applicability and limitations of the current approaches in disease modeling for AD. Also, we attempted to provide key suggestions for the best-fit model to evaluate potential therapies, which might improve therapy translation from preclinical studies to patients with AD.
Collapse
Affiliation(s)
- Ansab Akhtar
- Department
of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, Dehradun 248007, India
| | - Shraddha M. Gupta
- Department
of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, Dehradun 248007, India
| | - Shubham Dwivedi
- Department
of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, Dehradun 248007, India
| | - Devendra Kumar
- Faculty
of Pharmacy, DIT University, Uttarakhand, Dehradun 248009, India
| | - Mohd. Farooq Shaikh
- Neuropharmacology
Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 47500, Malaysia
| | - Arvind Negi
- Department
of Bioproducts and Biosystems, Aalto University, FI-00076 Espoo, Finland
- E-mail:
| |
Collapse
|
163
|
Collins AE, Saleh TM, Kalisch BE. VANL-100 Attenuates Beta-Amyloid-Induced Toxicity in SH-SY5Y Cells. Int J Mol Sci 2022; 24:ijms24010442. [PMID: 36613883 PMCID: PMC9820495 DOI: 10.3390/ijms24010442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/16/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022] Open
Abstract
Antioxidants are being explored as novel therapeutics for the treatment of neurodegenerative diseases such as Alzheimer's disease (AD) through strategies such as chemically linking antioxidants to synthesize novel co-drugs. The main objective of this study was to assess the cytoprotective effects of the novel antioxidant compound VANL-100 in a cellular model of beta-amyloid (Aβ)-induced toxicity. The cytotoxic effects of Aβ in the presence and absence of all antioxidant compounds were measured using the 3-(4,5-dimethylthiazol-2-yl)2-5-diphenyl-2H-tetrazolium bromide (MTT) assay in SH-SY5Y cells in both pre-treatment and co-treatment experiments. In pre-treatment experiments, VANL-100, or one of its parent compounds, naringenin (NAR), alpha-lipoic acid (ALA), or naringenin + alpha-lipoic acid (NAR + ALA), was administrated 24 h prior to an additional 24-h incubation with 20 μM non-fibril or fibril Aβ25-35. Co-treatment experiments consisted of simultaneous treatment with Aβ and antioxidants. Pre-treatment and co-treatment with VANL-100 significantly attenuated Aβ-induced cell death. There were no significant differences between the protective effects of VANL-100, NAR, ALA, and NAR + ALA with either form of Aβ, or in the effect of VANL-100 between 24-h pre-treatment and co-treatment. These results demonstrate that the novel co-drug VANL-100 is capable of eliciting cytoprotective effects against Aβ-induced toxicity.
Collapse
|
164
|
Ali A, Yu L, Kousar S, Khalid W, Maqbool Z, Aziz A, Arshad MS, Aadil RM, Trif M, Riaz S, Shaukat H, Manzoor MF, Qin H. Crocin: Functional characteristics, extraction, food applications and efficacy against brain related disorders. Front Nutr 2022; 9:1009807. [PMID: 36583211 PMCID: PMC9792498 DOI: 10.3389/fnut.2022.1009807] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
Crocin is a bioactive compound that naturally occurs in some medicinal plants, especially saffron and gardenia fruit. Different conventional and novel methods are used for its extraction. Due to some control conditions, recent methods such as ultrasonic extraction, supercritical fluid extraction, enzyme-associated extraction, microwave extraction, and pulsed electric field extraction are widely used because these methods give more yield and efficiency. Crocin is incorporated into different food products to make functional foods. However, it can also aid in the stability of food products. Due to its ability to protect against brain diseases, the demand for crocin has been rising in the pharmaceutical industry. It also contain antioxidant, anti-inflammatory, anticancer and antidepressant qualities. This review aims to describe crocin and its role in developing functional food, extraction, and bioavailability in various brain-related diseases. The results of the literature strongly support the importance of crocin against various diseases and its use in making different functional foods.
Collapse
Affiliation(s)
- Anwar Ali
- Xiangya School of Public Health, Central South University, Changsha, China
| | - Liang Yu
- Department of Research and Development Office, Hunan First Normal University, Changsha, China,*Correspondence: Liang Yu
| | - Safura Kousar
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Waseem Khalid
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Zahra Maqbool
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Afifa Aziz
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Sajid Arshad
- Department of Food Science, Government College University Faisalabad, Faisalabad, Pakistan
| | - Rana Muhammad Aadil
- National Institute of Food Science and Technology, University of Agriculture, Faisalabad, Pakistan
| | - Monica Trif
- Food Research Department, Centre for Innovative Process Engineering, Syke, Germany
| | - Sakhawat Riaz
- Department of Home Economics, Government College University, Faisalabad, Pakistan,Food and Nutrition Society, Gilgit Baltistan, Pakistan
| | - Horia Shaukat
- Xiangya School of Public Health, Central South University, Changsha, China
| | - Muhammad Faisal Manzoor
- Guangdong Provincial Key Laboratory of Intelligent Food Manufacturing, Foshan University, Foshan, China,School of Food Science and Engineering, South China University of Technology, Guangzhou, China,Muhammad Faisal Manzoor
| | - Hong Qin
- Xiangya School of Public Health, Central South University, Changsha, China,Hong Qin
| |
Collapse
|
165
|
Lu X, Qin N, Liu Y, Du C, Feng F, Liu W, Chen Y, Sun H. Design, synthesis, and biological evaluation of aromatic tertiary amine derivatives as selective butyrylcholinesterase inhibitors for the treatment of Alzheimer's disease. Eur J Med Chem 2022; 243:114729. [DOI: 10.1016/j.ejmech.2022.114729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/27/2022]
|
166
|
Yan N, Shi XL, Tang LQ, Wang DF, Li X, Liu C, Liu ZP. Synthesis and biological evaluation of thieno[3,2- c]pyrazol-3-amine derivatives as potent glycogen synthase kinase 3β inhibitors for Alzheimer's disease. J Enzyme Inhib Med Chem 2022; 37:1724-1736. [PMID: 35698879 PMCID: PMC9225722 DOI: 10.1080/14756366.2022.2086867] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Glycogen synthase kinase 3β (GSK-3β) catalyses the hyperphosphorylation of tau protein in the Alzheimer's disease (AD) pathology. A series of novel thieno[3,2-c]pyrazol-3-amine derivatives were designed and synthesised and evaluated as potential GSK-3β inhibitors by structure-guided drug rational design approach. The thieno[3,2-c]pyrazol-3-amine derivative 16b was identified as a potent GSK-3β inhibitor with an IC50 of 3.1 nM in vitro and showed accepted kinase selectivity. In cell levels, 16b showed no toxicity on the viability of SH-SY5Y cells at the concentration up to 50 μM and targeted GSK-3β with the increased phosphorylated GSK-3β at Ser9. Western blot analysis indicated that 16b decreased the phosphorylated tau at Ser396 in a dose-dependent way. Moreover, 16b effectively increased expressions of β-catenin as well as the GAP43, N-myc, and MAP-2, and promoted the differentiated neuronal neurite outgrowth. Therefore, the thieno[3,2-c]pyrazol-3-amine derivative 16b could serve as a promising GSK-3β inhibitor for the treatment of AD.
Collapse
Affiliation(s)
- Ning Yan
- Institute of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Xiao-Long Shi
- Institute of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Long-Qian Tang
- Institute of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - De-Feng Wang
- Institute of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Xun Li
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, PR China
| | - Chao Liu
- Institute of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Zhao-Peng Liu
- Institute of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| |
Collapse
|
167
|
Cheng X, Tan Y, Li H, Huang J, Zhao D, Zhang Z, Yi M, Zhu L, Hui S, Yang J, Peng W. Fecal 16S rRNA sequencing and multi-compartment metabolomics revealed gut microbiota and metabolites interactions in APP/PS1 mice. Comput Biol Med 2022; 151:106312. [PMID: 36417828 DOI: 10.1016/j.compbiomed.2022.106312] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/27/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Alzheimer's disease is a significant public health issue. Recent studies have shown that the gut microbiota plays a vital role in the onset and development of Alzheimer's disease. However, the potential role of the gut microbiota and the associated metabolic characteristics require further elucidation. METHODS The gut microbial compositions of APP/PS1 mice were analyzed using 16S rRNA gene sequencing. Metabolomics was used to characterize changes in metabolic profiles in feces, serum, and cortex. A multi-omics approach investigated the potential associations between gut microbes and metabolites. RESULTS The gut microbiota composition was markedly different between APP/PS1 mice and normal mice. Metabolomic analysis identified 253 fecal metabolites, 16 serum metabolites, and 123 cortical metabolites that were differentially abundant in APP/PS1 that may be potential biomarkers of AD. Nearly half of these metabolites were lipids. A combined analysis of the three sample types showed a correlation between fecal fatty acids and glycerolipids, serum glycerophospholipids, and cortical fatty acids. Furthermore, our study showed that Marinifilaceae and Akkermansiaceae were closely related to these lipids and lipid-like molecules, particularly fatty acids and glycerophospholipids. CONCLUSION Our study highlighted the interactions between the gut microbiome and the fecal, serum, and cortical metabolomes. This interaction provides a new direction for further exploring the link between gut microbiota composition and metabolism in Alzheimer's disease.
Collapse
Affiliation(s)
- Xin Cheng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; National Clinical Research Center for Mental Disorder, Changsha, 410011, China
| | - Yejun Tan
- School of Mathematics, University of Minnesota Twin Cities, Minneapolis, 55455, MN, USA
| | - Hongli Li
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; National Clinical Research Center for Mental Disorder, Changsha, 410011, China
| | - Jianhua Huang
- Hunan Academy of Chinese Medicine, Changsha, 410013, China
| | - Di Zhao
- Hunan Academy of Chinese Medicine, Changsha, 410013, China
| | - Zheyu Zhang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; National Clinical Research Center for Mental Disorder, Changsha, 410011, China
| | - Min Yi
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; National Clinical Research Center for Mental Disorder, Changsha, 410011, China
| | - Lemei Zhu
- Academician Workstation, Changsha Medical University, Changsha, 410219, China
| | - Shan Hui
- Department of Geratology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, 410005, China
| | - Jingjing Yang
- Teaching and Research Section of Clinical Nursing, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Weijun Peng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China; National Clinical Research Center for Mental Disorder, Changsha, 410011, China.
| |
Collapse
|
168
|
Ahmad F, Sachdeva P. Critical appraisal on mitochondrial dysfunction in Alzheimer's disease. Aging Med (Milton) 2022; 5:272-280. [PMID: 36606272 PMCID: PMC9805294 DOI: 10.1002/agm2.12217] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 01/09/2023] Open
Abstract
It is widely recognized that Alzheimer's disease (AD) is a common type of progressive neurodegenerative disorder that results in cognitive impairment over time. Approximately 152 million cases of AD are predicted to be reported by 2050. Amyloid plaques and tau proteins are two major hallmarks of AD which can be seen under electron microscope. Mitochondria plays a vital role in the pathogenesis of AD and mitochondria disruption leads to mitochondrial DNA (mtDNA) dysfunction, alteration of mitochondria dependent Ca2+ homeostasis, copper dysfunction, immune cell dysfunction, etc. In this review, we try to cover all the mechanisms related with mitochondrial dysfunction and mitochondrial pathogenesis that may help us to better understand AD as well as open a new era for therapeutic target of AD and treat this progressive disease.
Collapse
Affiliation(s)
- Faizan Ahmad
- Department of Medical Elementology and ToxicologyJamia Hamdard UniversityDelhiIndia
| | - Punya Sachdeva
- Amity Institute of Neuropsychology and NeurosciencesAmity UniversityNoidaUttar PradeshIndia
| |
Collapse
|
169
|
Moreira NCDS, Tamarozzi ER, Lima JEBDF, Piassi LDO, Carvalho I, Passos GA, Sakamoto-Hojo ET. Novel Dual AChE and ROCK2 Inhibitor Induces Neurogenesis via PTEN/AKT Pathway in Alzheimer's Disease Model. Int J Mol Sci 2022; 23:ijms232314788. [PMID: 36499116 PMCID: PMC9737254 DOI: 10.3390/ijms232314788] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/07/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive and complex neurodegenerative disease. Acetylcholinesterase inhibitors (AChEIs) are a major class of drugs used in AD therapy. ROCK2, another promising target for AD, has been associated with the induction of neurogenesis via PTEN/AKT. This study aimed to characterize the therapeutic potential of a novel donepezil-tacrine hybrid compound (TA8Amino) to inhibit AChE and ROCK2 protein, leading to the induction of neurogenesis in SH-SY5Y cells. Experiments were carried out with undifferentiated and neuron-differentiated SH-SY5Y cells submitted to treatments with AChEIs (TA8Amino, donepezil, and tacrine) for 24 h or 7 days. TA8Amino was capable of inhibiting AChE at non-cytotoxic concentrations after 24 h. Following neuronal differentiation for 7 days, TA8Amino and donepezil increased the percentage of neurodifferentiated cells and the length of neurites, as confirmed by β-III-tubulin and MAP2 protein expression. TA8Amino was found to participate in the activation of PTEN/AKT signaling. In silico analysis showed that TA8Amino can stably bind to the active site of ROCK2, and in vitro experiments in SH-SY5Y cells demonstrate that TA8Amino significantly reduced the expression of ROCK2 protein, contrasting with donepezil and tacrine. Therefore, these results provide important information on the mechanism underlying the action of TA8Amino with regard to multi-target activities.
Collapse
Affiliation(s)
| | - Elvira Regina Tamarozzi
- Department of Biotechnology, School of Arts, Sciences and Humanities—USP, São Paulo 03828-000, Brazil
| | | | - Larissa de Oliveira Piassi
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo—USP, Ribeirão Preto 14049-900, Brazil
| | - Ivone Carvalho
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo—USP, Ribeirão Preto 14040-900, Brazil
| | - Geraldo Aleixo Passos
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo—USP, Ribeirão Preto 14049-900, Brazil
- Laboratory of Genetics and Molecular Biology, Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14049-900, Brazil
| | - Elza Tiemi Sakamoto-Hojo
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo—USP, Ribeirão Preto 14049-900, Brazil
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo—USP, Ribeirão Preto 14040-901, Brazil
- Correspondence: ; Tel.: +55-16-3315-3827
| |
Collapse
|
170
|
Li ZL, Ma HT, Wang M, Qian YH. Research trend of microbiota-gut-brain axis in Alzheimer's disease based on CiteSpace (2012-2021): A bibliometrics analysis of 608 articles. Front Aging Neurosci 2022; 14:1036120. [PMID: 36483116 PMCID: PMC9724362 DOI: 10.3389/fnagi.2022.1036120] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/03/2022] [Indexed: 05/12/2025] Open
Abstract
BACKGROUND Recently, research on the microbiota-gut-brain axis (MGBA) has received increasing attention, and the number of studies related to Alzheimer's disease (AD) has increased rapidly, but there is currently a lack of summary of MGBA in AD. OBJECTIVE To capture research hotspots, grasp the context of disciplinary research, and explore future research development directions. METHODS In the core dataset of Web of Science, documents are searched according to specific subject words. CiteSpace software is used to perform statistical analysis on measurement indicators such as the number of published papers, publishing countries, institutions, subject areas, authors, cocited journals, and keywords, and to visualize of a network of relevant content elements. RESULTS The research of MGBA in AD has shown an upward trend year by year, and the cooperation between countries is relatively close, and mainly involves the intersection of neuroscience, pharmacy, and microbiology. This research focuses on the relationship between MGBA and AD symptoms. Keyword hotspots are closely related to new technologies. Alzheimer's disease, anterior cingulate cortex, inflammatory degeneration, dysbiosis, and other research are the focus of this field. CONCLUSION The study revealed that the research and development of MGBA in AD rapidly progressed, but no breakthrough has been made in the past decade, it still needs to be closely combined with multidisciplinary technology to grasp the frontier hotspots. Countries should further strengthen cooperation, improve the disciplinary system, and increase the proportion of empirical research in all research.
Collapse
Affiliation(s)
- Zi-Long Li
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institute of Neuroscience, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Hao-Tian Ma
- College of Forensic Science, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Meng Wang
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institute of Neuroscience, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Yi-Hua Qian
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Institute of Neuroscience, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| |
Collapse
|
171
|
Lengerli D, Ibis K, Nural Y, Banoglu E. The 1,2,3-triazole 'all-in-one' ring system in drug discovery: a good bioisostere, a good pharmacophore, a good linker, and a versatile synthetic tool. Expert Opin Drug Discov 2022; 17:1209-1236. [PMID: 36164263 DOI: 10.1080/17460441.2022.2129613] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION The 1,2,3-triazole ring occupies an important space in medicinal chemistry due to its unique structural properties, synthetic versatility and pharmacological potential making it a critical scaffold. Since it is readily available through click chemistry for creating compound collections against various diseases, it has become an emerging area of interest for medicinal chemists. AREAS COVERED This review article addresses the unique properties of the1,2,3-triazole nucleus as an intriguing ring system in drug discovery while focusing on the most recent medicinal chemistry strategies exploited for the design and development of 1,2,3-triazole analogs as inhibitors of various biological targets. EXPERT OPINION Evidently, the 1,2,3-triazole ring with unique structural features has enormous potential in drug design against various diseases as a pharmacophore, a bioisoster or a structural platform. The most recent evidence indicates that it may be more emerging in drug molecules in near future along with an increasing understanding of its prominent roles in drug structures. The synthetic feasibility and versatility of triazole chemistry make it certainly ideal for creating compound libraries for more constructive structure-activity relationship studies. However, more comparative and target-specific studies are needed to gain a deeper understanding of the roles of the 1,2,3-triazole ring in molecular recognition.[Figure: see text].
Collapse
Affiliation(s)
- Deniz Lengerli
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| | - Kübra Ibis
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| | - Yahya Nural
- Department of Analytical Chemistry, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Erden Banoglu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| |
Collapse
|
172
|
Li H, Zha S, Li H, Liu H, Wong KL, All AH. Polymeric Dendrimers as Nanocarrier Vectors for Neurotheranostics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203629. [PMID: 36084240 DOI: 10.1002/smll.202203629] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/01/2022] [Indexed: 06/15/2023]
Abstract
Dendrimers are polymers with well-defined 3D branched structures that are vastly utilized in various neurotheranostics and biomedical applications, particularly as nanocarrier vectors. Imaging agents can be loaded into dendrimers to improve the accuracy of diagnostic imaging processes. Likewise, combining pharmaceutical agents and anticancer drugs with dendrimers can enhance their solubility, biocompatibility, and efficiency. Practically, by modifying ligands on the surface of dendrimers, effective therapeutic and diagnostic platforms can be constructed and implemented for targeted delivery. Dendrimer-based nanocarriers also show great potential in gene delivery. Since enzymes can degrade genetic materials during their blood circulation, dendrimers exhibit promising packaging and delivery alternatives, particularly for central nervous system (CNS) treatments. The DNA and RNA encapsulated in dendrimers represented by polyamidoamine that are used for targeted brain delivery, via chemical-structural adjustments and appropriate generation, significantly improve the correlation between transfection efficiency and cytotoxicity. This article reports a comprehensive review of dendrimers' structures, synthesis processes, and biological applications. Recent progress in diagnostic imaging processes and therapeutic applications for cancers and other CNS diseases are presented. Potential challenges and future directions in the development of dendrimers, which provide the theoretical basis for their broader applications in healthcare, are also discussed.
Collapse
Affiliation(s)
- Hengde Li
- Department of Chemistry, Hong Kong Baptist University, 224 Waterloo Road, Kowloon, Hong Kong SAR, P. R. China
| | - Shuai Zha
- Department of Chemistry, Hong Kong Baptist University, 224 Waterloo Road, Kowloon, Hong Kong SAR, P. R. China
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, P. R. China
| | - Haolan Li
- Department of Chemistry, Hong Kong Baptist University, 224 Waterloo Road, Kowloon, Hong Kong SAR, P. R. China
| | - Haitao Liu
- Department of Chemistry, Hong Kong Baptist University, 224 Waterloo Road, Kowloon, Hong Kong SAR, P. R. China
| | - Ka-Leung Wong
- Department of Chemistry, Hong Kong Baptist University, 224 Waterloo Road, Kowloon, Hong Kong SAR, P. R. China
| | - Angelo H All
- Department of Chemistry, Hong Kong Baptist University, 224 Waterloo Road, Kowloon, Hong Kong SAR, P. R. China
| |
Collapse
|
173
|
Neganova ME, Aleksandrova YR, Sukocheva OA, Klochkov SG. Benefits and limitations of nanomedicine treatment of brain cancers and age-dependent neurodegenerative disorders. Semin Cancer Biol 2022; 86:805-833. [PMID: 35779712 DOI: 10.1016/j.semcancer.2022.06.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/25/2022] [Accepted: 06/25/2022] [Indexed: 02/07/2023]
Abstract
The treatment of central nervous system (CNS) malignancies, including brain cancers, is limited by a number of obstructions, including the blood-brain barrier (BBB), the heterogeneity and high invasiveness of tumors, the inaccessibility of tissues for early diagnosis and effective surgery, and anti-cancer drug resistance. Therapies employing nanomedicine have been shown to facilitate drug penetration across the BBB and maintain biodistribution and accumulation of therapeutic agents at the desired target site. The application of lipid-, polymer-, or metal-based nanocarriers represents an advanced drug delivery system for a growing group of anti-cancer chemicals. The nanocarrier surface is designed to contain an active ligand (cancer cell marker or antibody)-binding structure which can be modified to target specific cancer cells. Glioblastoma, ependymoma, neuroblastoma, medulloblastoma, and primary CNS lymphomas were recently targeted by easily absorbed nanocarriers. The metal- (such as transferrin drug-loaded systems), polymer- (nanocapsules and nanospheres), or lipid- (such as sulfatide-containing nanoliposomes)-based nano-vehicles were loaded with apoptosis- and/or ferroptosis-stimulating agents and demonstrated promising anti-cancer effects. This review aims to discuss effective nanomedicine approaches designed to overcome the current limitations in the therapy of brain cancers and age-dependent neurodegenerative disorders. To accent current obstacles for successful CNS-based cancer therapy, we discuss nanomedicine perspectives and limitations of nanodrug use associated with the specificity of nervous tissue characteristics and the effects nanocarriers have on cognition.
Collapse
Affiliation(s)
- Margarita E Neganova
- Institute of Physiologically Active Compounds of the Russian Academy of Sciences, 1, Severnii pr., Chernogolovka 142432, Russia
| | - Yulia R Aleksandrova
- Institute of Physiologically Active Compounds of the Russian Academy of Sciences, 1, Severnii pr., Chernogolovka 142432, Russia
| | - Olga A Sukocheva
- School of Health Sciences, Flinders University of South Australia, Bedford Park, SA 5042, Australia.
| | - Sergey G Klochkov
- Institute of Physiologically Active Compounds of the Russian Academy of Sciences, 1, Severnii pr., Chernogolovka 142432, Russia
| |
Collapse
|
174
|
Ferreira I, Rauter AP, Bandarra NM. Marine Sources of DHA-Rich Phospholipids with Anti-Alzheimer Effect. Mar Drugs 2022; 20:662. [PMID: 36354985 PMCID: PMC9695993 DOI: 10.3390/md20110662] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 07/29/2023] Open
Abstract
Alzheimer's disease (AD) is a complex and progressive disease, which affects millions of people around the world. Despite the many efforts over the years to find efficient therapeutics, there is no cure yet. Nonetheless, many compounds have been proven to decrease Alzheimer's symptoms. After a short overview of the hypotheses considered in AD drug development and the drugs approved for AD treatment, which lead to symptom release, we focus on the valorization of natural marine sources that decrease AD symptoms, particularly on docosahexaenoic acid (DHA), an important component in membrane phospholipids and the most abundant n-3 polyunsaturated fatty acids (PUFA) found in gray matter of the brain and in retina and on the DHA-containing phospholipids (DHA-PLs) present in marine sources, namely fish, krill, mollusks and in fisheries and aquaculture by-products. DHA-PLs' bioactivities are presented, namely their properties in anti-neurodegeneration, neuroinflammation, as anticancer agents, as well as their benefits to obesity and visual problems. Fisheries and aquaculture by-products are also highlighted as they have a high content of DHA and DHA-rich phospholipids, can be extracted by green methodologies and should be considered in a circular economy for a healthy sustainable future.
Collapse
Affiliation(s)
- Inês Ferreira
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
- Division of Aquaculture, Upgrading and Bioprospecting, Portuguese Institute of the Sea and Atmosphere, 1495-165 Lisboa, Portugal
| | - Amélia P. Rauter
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Narcisa M. Bandarra
- Division of Aquaculture, Upgrading and Bioprospecting, Portuguese Institute of the Sea and Atmosphere, 1495-165 Lisboa, Portugal
- CIIMAR, Interdisciplinary Centre of Marine and Environmental Research, University of Porto, 4050-123 Porto, Portugal
| |
Collapse
|
175
|
Liu X, Ye M, Ma L. The emerging role of autophagy and mitophagy in tauopathies: From pathogenesis to translational implications in Alzheimer's disease. Front Aging Neurosci 2022; 14:1022821. [PMID: 36325189 PMCID: PMC9618726 DOI: 10.3389/fnagi.2022.1022821] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/27/2022] [Indexed: 09/15/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease, affecting more than 55 million individuals worldwide in 2021. In addition to the "amyloid hypothesis," an increasing number of studies have demonstrated that phosphorylated tau plays an important role in AD pathogenesis. Both soluble tau oligomers and insoluble tau aggregates in the brain can induce structural and functional neuronal damage through multiple pathways, eventually leading to memory deficits and neurodegeneration. Autophagy is an important cellular response to various stress stimuli and can generally be categorized into non-selective and selective autophagy. Recent studies have indicated that both types of autophagy are involved in AD pathology. Among the several subtypes of selective autophagy, mitophagy, which mediates the selective removal of mitochondria, has attracted increasing attention because dysfunctional mitochondria have been suggested to contribute to tauopathies. In this review, we summarize the latest findings on the bidirectional association between abnormal tau proteins and defective autophagy, as well as mitophagy, which might constitute a vicious cycle in the induction of neurodegeneration. Neuroinflammation, another important feature in the pathogenesis and progression of AD, has been shown to crosstalk with autophagy and mitophagy. Additionally, we comprehensively discuss the relationship between neuroinflammation, autophagy, and mitophagy. By elucidating the underlying molecular mechanisms governing these pathologies, we highlight novel therapeutic strategies targeting autophagy, mitophagy and neuroinflammation, such as those using rapamycin, urolithin, spermidine, curcumin, nicotinamide, and actinonin, for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Xiaolan Liu
- Wuhan Mental Health Center, Wuhan, China
- Wuhan Hospital for Psychotherapy, Wuhan, China
| | - Meng Ye
- Wuhan Mental Health Center, Wuhan, China
- Wuhan Hospital for Psychotherapy, Wuhan, China
| | - Liang Ma
- Wuhan Mental Health Center, Wuhan, China
- Wuhan Hospital for Psychotherapy, Wuhan, China
| |
Collapse
|
176
|
Shiravandi A, Yari F, Tofigh N, Kazemi Ashtiani M, Shahpasand K, Ghanian MH, Shekari F, Faridbod F. Earlier Detection of Alzheimer's Disease Based on a Novel Biomarker cis P-tau by a Label-Free Electrochemical Immunosensor. BIOSENSORS 2022; 12:879. [PMID: 36291017 PMCID: PMC9599477 DOI: 10.3390/bios12100879] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/01/2022] [Accepted: 10/07/2022] [Indexed: 06/16/2023]
Abstract
Early detection of cis phosphorylated tau (cis P-tau) may help as an effective treatment to control the progression of Alzheimer's disease (AD). Recently, we introduced for the first time a monoclonal antibody (mAb) with high affinity against cis P-tau. In this study, the cis P-tau mAb was utilized to develop a label-free immunosensor. The antibody was immobilized onto a gold electrode and the electrochemical responses to the analyte were acquired by electrochemical impedance spectroscopy (EIS), cyclic voltammetry (CV), and differential pulse voltammetry (DPV). The immunosensor was capable of selective detection of cis P-tau among non-specific targets like trans P-tau and major plasma proteins. A wide concentration range (10 × 10-14 M-3.0 × 10-9 M) of cis P-tau was measured in PBS and human serum matrices with a limit of detection of 0.02 and 0.05 pM, respectively. Clinical applicability of the immunosensor was suggested by its long-term storage stability and successful detection of cis P-tau in real samples of cerebrospinal fluid (CSF) and blood serum collected from human patients at different stages of AD. These results suggest that this simple immunosensor may find great application in clinical settings for early detection of AD which is an unmet urgent need in today's healthcare services.
Collapse
Affiliation(s)
- Ayoub Shiravandi
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran
| | - Farzaneh Yari
- Center of Excellence in Electrochemistry, School of Chemistry, College of Science, University of Tehran, Tehran P.O. Box 14155-6455, Iran
| | - Nahid Tofigh
- Laboratory of Neuro-Organic Chemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran 1417935840, Iran
| | - Mohammad Kazemi Ashtiani
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran
| | - Koorosh Shahpasand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran
| | - Mohammad-Hossein Ghanian
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran
| | - Faezeh Shekari
- Advanced Therapy Medicinal Product Technology Development Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran
| | - Farnoush Faridbod
- Center of Excellence in Electrochemistry, School of Chemistry, College of Science, University of Tehran, Tehran P.O. Box 14155-6455, Iran
| |
Collapse
|
177
|
BozbeyMerde İ, Önel GT, Türkmenoğlu B, Gürsoy Ş, Dilek E. (
p
‐Chlorophenyl)‐3(2
H
)pyridazinone Derivatives: Synthesis,
in Silico
, and AChE/BChE Inhibitory Activity. ChemistrySelect 2022. [DOI: 10.1002/slct.202202446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- İrem BozbeyMerde
- Department of Pharmaceutical Chemistry Faculty of Pharmacy Erzincan Binali Yıldırım University Yalnızbağ 24002 Erzincan Turkey
| | - Gülce Taşkor Önel
- Department of Analytical Chemistry Faculty of Pharmacy Erzincan Binali Yıldırım University Yalnızbağ 24002 Erzincan Turkey
| | - Burçin Türkmenoğlu
- Department of Analytical Chemistry Faculty of Pharmacy Erzincan Binali Yıldırım University Yalnızbağ 24002 Erzincan Turkey
| | - Şule Gürsoy
- Department of Biochemistry Faculty of Pharmacy Erzincan Binali Yıldırım University Yalnızbağ 24002 Erzincan Turkey
| | - Esra Dilek
- Department of Biochemistry Faculty of Pharmacy Erzincan Binali Yıldırım University Yalnızbağ 24002 Erzincan Turkey
| |
Collapse
|
178
|
Ravi R, Zeyaullah M, Ghosh S, Khan Warsi M, Baweja R, AlShahrani AM, Mishra A, Ahmad R. Use of gold nanoparticle-silibinin conjugates: A novel approach against lung cancer cells. Front Chem 2022; 10:1018759. [PMID: 36311430 PMCID: PMC9606463 DOI: 10.3389/fchem.2022.1018759] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 09/23/2022] [Indexed: 08/11/2023] Open
Abstract
Lung cancer presents one of the most challenging carcinomas with meager 5-year survival rates (less than 20%), high metastasis and high recurrence due to chemo- and radio- resistance. An alternative or complementation to existing prognosis modalities is the use of phytochemicals such as silibinin, which targets essential cytokines, angiogenic factors and transcription factors for a profound anti-tumor effect. However, the problems of low solubility in an aqueous physiological environment, poor penetration, high metabolism and rapid systemic clearance limit the therapeutic use of silibinin. Conjugation of gold nanoparticles (GNPs) with silibinin may overcome the above challenges along with distinct advantages of biocompatibility, optical properties for monitoring and causation of cytotoxicity in cancer cells. The current study thus aims to develop silibinin conjugated gold nanoparticles (Sb-GNPs) with pH responsive release in the cancer microenvironment, optimizing several parameters for its higher activity and further evaluate the nanoplatform for their efficacy in inducing cell death in vitro against A549 lung cancer cells. GNPs was synthesized using trisodium citrate dihydrate as the reducing agent and further used for the conjugation of silibinin. The synthesized GNPs were found to be monodispersed and spherical in shape. The silibinin was successfully conjugated with gold nanoparticles and long-term stability of GNPs and Sb-GNPs nanoconjugates in suspension phase was confirmed by FTIR and DLS. Anticancer properties of Sb-GNPs were confirmed by different assay using MTT, Trypan blue dye exclusion assay and cell cycle analysis assay. After conjugation of silibinin with GNPs, the efficacy of silibinin increased 4-5 times in killing the cancer cells. This is the first report on using silibinin gold nanoconjugate system for lung cancer therapy with promising future applications.
Collapse
Affiliation(s)
- Rangnath Ravi
- Department of Chemistry, Shivaji College, University of Delhi, New Delhi, India
| | - Md. Zeyaullah
- Department of Basic Medical Science, College of Applied Medical Sciences, King Khalid University (KKU), Khamis Mushayt Campus, Abha, Saudi Arabia
| | - Shubhrima Ghosh
- Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Mohiuddin Khan Warsi
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Renu Baweja
- Department of Biochemistry, Shivaji College, University of Delhi, New Delhi, India
| | - Abdullah M. AlShahrani
- Department of Basic Medical Science, College of Applied Medical Sciences, King Khalid University (KKU), Khamis Mushayt Campus, Abha, Saudi Arabia
| | - Abhijeet Mishra
- Department of Biochemistry, Shivaji College, University of Delhi, New Delhi, India
| | - Razi Ahmad
- Department of Chemistry, Indian Institute of Technology Delhi, New Delhi, India
- Quality and Research Department, Anantaa GSK Innovations Pvt Ltd., DLF Industrial Area, Faridabad, India
| |
Collapse
|
179
|
Li X, Feng X, Sun X, Hou N, Han F, Liu Y. Global, regional, and national burden of Alzheimer's disease and other dementias, 1990-2019. Front Aging Neurosci 2022; 14:937486. [PMID: 36299608 PMCID: PMC9588915 DOI: 10.3389/fnagi.2022.937486] [Citation(s) in RCA: 300] [Impact Index Per Article: 100.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND With the increase in the aging population worldwide, Alzheimer's disease has become a rapidly increasing public health concern. Monitoring the dementia disease burden will support health development strategies by providing scientific data. METHODS Based on the data obtained from the 2019 Global Burden of Disease (GBD) database, the numbers and age-standardized rates (ASRs) of incidence, prevalence, death, and disability-adjusted life-years (DALYs) of Alzheimer's disease and other dementias from 1990 to 2019 were analyzed. Calculated estimated annual percentage changes (EAPCs) and Joinpoint regression analyses were performed to evaluate the trends during this period. We also evaluated the correlations between the epidemiology and the sociodemographic index (SDI), an indicator to evaluate the level of social development in a country or region considering the education rate, economic situation, and total fertility rate. RESULTS From 1990 to 2019, the incidence and prevalence of Alzheimer's disease and other dementias increased by 147.95 and 160.84%, respectively. The ASR of incidence, prevalence, death, and DALYs in both men and women consistently increased over the study period. All the ASRs in women were consistently higher than those in men, but the increases were more pronounced in men. In addition, the ASRs of incidence, prevalence, and DALYs were positively correlated with the SDI. Moreover, the proportion of patients over 70 years old with dementia was also positively correlated with the SDI level. Smoking was a major risk factor for the disease burden of dementia in men, while obesity was the major risk factor for women. CONCLUSION From 1990 to 2019, the Alzheimer's disease burden increased worldwide. This trend was more serious in high-SDI areas, especially among elderly populations in high-SDI areas, who should receive additional attention. Policy-makers should take steps to reverse this situation. Notably, women were at a higher risk for the disease, but the risk in men showed a faster increase. We should give attention to the aging population, attach importance to interventions targeting dementia risk factors, and formulate action plans to address the increasing incidence of dementia.
Collapse
Affiliation(s)
- Xue Li
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
- Department of Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xiaojin Feng
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Department of Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Department of Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Department of Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Fang Han
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yongping Liu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Department of Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| |
Collapse
|
180
|
Shi B, Zhao J, Xu Z, Chen C, Xu L, Xu C, Sun M, Kuang H. Chiral Nanoparticles Force Neural Stem Cell Differentiation to Alleviate Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202475. [PMID: 36008133 PMCID: PMC9561871 DOI: 10.1002/advs.202202475] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/08/2022] [Indexed: 06/04/2023]
Abstract
The differentiation of neural stem cells via nanomaterials has attracted attention and has become a potential tool. However, the chirality effect in neural stem cell differentiation has not been investigated. Here, this study shows that chiral nanoparticles (NPs) with strong chirality can efficiently accelerate the differentiation of mouse neural stem cells (NSCs) into neurons under near-infrared (NIR) light illumination. L-type NPs are 1.95 times greater than D-type NPs in promoting NSCs differentiation due to their 1.47-fold endocytosis efficiency. Whole gene expression map analysis reveals that circularly polarized light illumination and chiral NPs irradiation significantly upregulate Map2, Yap1, and Taz genes, resulting in mechanical force, cytoskeleton protein action, and accelerated NSCs differentiation. In vivo experiments show that successful differentiation can further alleviate symptoms in Alzheimer's disease mice. Moreover, the clearance of L-type NPs on amyloid and hyperphosphorylated p-tau protein reachs 68.24% and 66.43%, respectively, under the synergy of NIR irradiation. The findings suggest that strong chiral nanomaterials may have advantages in guiding cell development and can be used in biomedicine.
Collapse
Affiliation(s)
- Baimei Shi
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Jing Zhao
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Zhuojia Xu
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Chen Chen
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Liguang Xu
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Chuanlai Xu
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Maozhong Sun
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Hua Kuang
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| |
Collapse
|
181
|
Sun ZD, Hu JX, Wu JR, Zhou B, Huang YP. Toxicities of amyloid-beta and tau protein are reciprocally enhanced in the Drosophila model. Neural Regen Res 2022; 17:2286-2292. [PMID: 35259851 PMCID: PMC9083152 DOI: 10.4103/1673-5374.336872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Extracellular aggregation of amyloid-beta (Aβ) and intracellular tau tangles are two major pathogenic hallmarks and critical factors of Alzheimer’s disease. A linear interaction between Aβ and tau protein has been characterized in several models. Aβ induces tau hyperphosphorylation through a complex mechanism; however, the master regulators involved in this linear process are still unclear. In our study with Drosophila melanogaster, we found that Aβ regulated tau hyperphosphorylation and toxicity by activating c-Jun N-terminal kinase. Importantly, Aβ toxicity was dependent on tau hyperphosphorylation, and flies with hypophosphorylated tau were insulated against Aβ-induced toxicity. Strikingly, tau accumulation reciprocally interfered with Aβ degradation and correlated with the reduction in mRNA expression of genes encoding Aβ-degrading enzymes, including dNep1, dNep3, dMmp2, dNep4, and dIDE. Our results indicate that Aβ and tau protein work synergistically to further accelerate Alzheimer’s disease progression and may be considered as a combined target for future development of Alzheimer’s disease therapeutics.
Collapse
Affiliation(s)
- Zhen-Dong Sun
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang Province, China
| | - Jia-Xin Hu
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang Province, China
| | - Jia-Rui Wu
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang Province, China
| | - Bing Zhou
- State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yun-Peng Huang
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang Province, China
| |
Collapse
|
182
|
Nose-to-Brain: The Next Step for Stem Cell and Biomaterial Therapy in Neurological Disorders. Cells 2022; 11:cells11193095. [PMID: 36231058 PMCID: PMC9564248 DOI: 10.3390/cells11193095] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/28/2022] [Accepted: 09/29/2022] [Indexed: 11/18/2022] Open
Abstract
Neurological disorders are a leading cause of morbidity worldwide, giving rise to a growing need to develop treatments to revert their symptoms. This review highlights the great potential of recent advances in cell therapy for the treatment of neurological disorders. Through the administration of pluripotent or stem cells, this novel therapy may promote neuroprotection, neuroplasticity, and neuroregeneration in lesion areas. The review also addresses the administration of these therapeutic molecules by the intranasal route, a promising, non-conventional route that allows for direct access to the central nervous system without crossing the blood–brain barrier, avoiding potential adverse reactions and enabling the administration of large quantities of therapeutic molecules to the brain. Finally, we focus on the need to use biomaterials, which play an important role as nutrient carriers, scaffolds, and immune modulators in the administration of non-autologous cells. Little research has been conducted into the integration of biomaterials alongside intranasally administered cell therapy, a highly promising approach for the treatment of neurological disorders.
Collapse
|
183
|
Han R, Wang M, Wang L, Zhang Y, Li X, Hou Y, Yan J, Pan X. GC/MS-Based Urine Metabolomics Study on the Ameliorative Effect of Xanthoceras sorbifolia Extract on Alzheimer's Disease in Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:3390034. [PMID: 36164398 PMCID: PMC9509262 DOI: 10.1155/2022/3390034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 08/25/2022] [Indexed: 11/18/2022]
Abstract
The cause of Alzheimer's disease, the most common type of dementia today, is still unclear, and in current research, there are no drugs that work relatively well. Therefore, the study for new drugs to treat Alzheimer's disease is an urgent research need. Research on the improvement of Alzheimer's disease with extracts of Xanthoceras sorbifolia has been increasing in recent years, but the mechanism is not fully understood. The experiments were conducted to validate the model and analyze the treatment effect through D-galactose and Aβ 25-35 induced dementia model mice, using the Morris water maze, to detect the learning behavior and brain tissue section to observe the hippocampal tissue structure of mice. We performed a nontargeted metabolomic analysis of the urine obtained from different groups of mice using gas chromatography-mass spectrometry. Fourteen potential biomarkers were identified in the mice's urine, outlining five metabolic pathways of interest. It was shown that the extracts of Xanthoceras sorbifolia may exert protective effects on mice in dementia models through energy metabolism, neuroinflammation, and antioxidants. This study reveals the potential pathogenesis of Alzheimer's disease and the possible therapeutic mechanism of Xanthoceras sorbifolia, suggests relevant biomarkers, and provides an additional basis for the clinical application of Xanthoceras sorbifolia.
Collapse
Affiliation(s)
- Rui Han
- School of Stomatology, Lanzhou University, Donggang Road No. 199, Lanzhou 730020, China
| | - Min Wang
- School of Basic Medical Sciences, Lanzhou University, Donggang Road No. 199, Lanzhou 730020, China
| | - Li Wang
- School of Stomatology, Lanzhou University, Donggang Road No. 199, Lanzhou 730020, China
| | - Yichen Zhang
- School of Stomatology, Lanzhou University, Donggang Road No. 199, Lanzhou 730020, China
| | - Xin Li
- School of Stomatology, Lanzhou University, Donggang Road No. 199, Lanzhou 730020, China
| | - Yijun Hou
- School of Stomatology, Lanzhou University, Donggang Road No. 199, Lanzhou 730020, China
| | - Jing Yan
- School of Stomatology, Lanzhou University, Donggang Road No. 199, Lanzhou 730020, China
| | - Xiaojing Pan
- School of Stomatology, Lanzhou University, Donggang Road No. 199, Lanzhou 730020, China
| |
Collapse
|
184
|
Esmer Yİ, Çınar E, Başaran E. Design, Docking, Synthesis and Biological Evaluation of Novel Nicotinohydrazone Derivatives as Potential Butyrylcholinesterase Enzyme Inhibitor. ChemistrySelect 2022. [DOI: 10.1002/slct.202202771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Yusuf İslam Esmer
- Department of Chemistry Graduate Education Institute Batman University 72060 Batman Turkey
| | - Ercan Çınar
- Department of Nursing, Faculty of Health Sciences Batman University 72060 Batman Turkey
| | - Eyüp Başaran
- Department of Chemistry and Chemical Processing Technologies, Vocational School of Technical Sciences Batman University 72060 Batman Turkey
| |
Collapse
|
185
|
Wang P, Yu L, Gong J, Xiong J, Zi S, Xie H, Zhang F, Mao Z, Liu Z, Kim JS. An Activity‐Based Fluorescent Probe for Imaging Fluctuations of Peroxynitrite (ONOO
−
) in the Alzheimer's Disease Brain. Angew Chem Int Ed Engl 2022; 61:e202206894. [DOI: 10.1002/anie.202206894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Indexed: 12/25/2022]
Affiliation(s)
- Pengzhan Wang
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules College of Chemistry and Chemical Engineering Hubei University Wuhan 430062 China
| | - Le Yu
- Department of Chemistry Korea University Seoul 02841 Korea
| | - Jiankang Gong
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules College of Chemistry and Chemical Engineering Hubei University Wuhan 430062 China
| | - Jianhua Xiong
- College of Chemistry and Molecular Sciences Wuhan University Wuhan Hubei 430072 China
| | - Soyu Zi
- Department of Chemistry Korea University Seoul 02841 Korea
| | - Hua Xie
- School of Water Resources and Hydropower Wuhan University Wuhan Hubei 430072 China
| | - Fan Zhang
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules College of Chemistry and Chemical Engineering Hubei University Wuhan 430062 China
| | - Zhiqiang Mao
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules College of Chemistry and Chemical Engineering Hubei University Wuhan 430062 China
- Department of Chemistry Korea University Seoul 02841 Korea
| | - Zhihong Liu
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules College of Chemistry and Chemical Engineering Hubei University Wuhan 430062 China
- College of Chemistry and Molecular Sciences Wuhan University Wuhan Hubei 430072 China
| | - Jong Seung Kim
- Department of Chemistry Korea University Seoul 02841 Korea
| |
Collapse
|
186
|
Exploring the Formation of Polymers with Anti-Amyloid Properties within the 2′3′-Dihydroxyflavone Autoxidation Process. Antioxidants (Basel) 2022; 11:antiox11091711. [PMID: 36139781 PMCID: PMC9495709 DOI: 10.3390/antiox11091711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/26/2022] [Accepted: 08/27/2022] [Indexed: 11/16/2022] Open
Abstract
Amyloid-β and α-synuclein aggregation into amyloid fibrils is linked to the onset and progression of Alzheimer’s and Parkinson’s diseases. While there are only a few disease-modifying drugs, it is essential to search for new, more effective ways to encounter these neurodegenerative diseases. Multiple research articles have shown that the autoxidation of flavone is a critical factor for activating the inhibitory potential against the protein aggregation. Despite this, the structure of the newly-formed inhibitors is unknown. In this research, we examined the autoxidation products of 2′,3′-dihydroxyflavone that were previously shown to possess one of the most prominent inhibitory effects against amyloid-β aggregation. Their analysis using HPLC suggested the formation of polymeric molecules that were isolated using a 3 kDa cut-off. These polymeric structures were indicated as the most potent inhibitors based on protein aggregation kinetics and AFM studies. This revelation was confirmed using MALDI-TOF and NMR. We also show that active molecules have a tendency to reduce the Amyloid-β and α-synuclein aggregates toxicity to SH-SY5Y cells.
Collapse
|
187
|
Fidan GS, Parlar S, Tarikogullari AH, Alptuzun V, Alpan AS. Design, synthesis, acetylcholinesterase, butyrylcholinesterase, and amyloid-β aggregation inhibition studies of substituted 4,4'-diimine/4,4'-diazobiphenyl derivatives. Arch Pharm (Weinheim) 2022; 355:e2200152. [PMID: 35976708 DOI: 10.1002/ardp.202200152] [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: 03/25/2022] [Revised: 07/01/2022] [Accepted: 07/23/2022] [Indexed: 11/11/2022]
Abstract
A series of 4,4'-diimine/4,4'-diazobiphenyl derivatives were designed, synthesized, and evaluated for their ability to inhibit both the acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) enzymes, as well as Aβ1-42 aggregation, in vitro. The AChE and BChE inhibition assays demonstrated that all compounds displayed moderate AChE inhibitory activity in the range of IC50 = 5.77-16.22 μM, while they displayed weak or no BChE inhibition. Among the title compounds, compound 2l, 4,4'-bis(quinolin-8-yldiazenyl)-1,1'-biphenyl, having a diazo-quinoline moiety demonstrated the most potent inhibition against AChE with an IC50 value of 5.77 μM. Furthermore, diazo derivatives 2d, 4,4'-bis[(4-methoxyphenyl)diazenyl]-1,1'-biphenyl, and 2i, 4,4'-bis(pyridin-3-yldiazenyl)-1,1'-biphenyl, provided better potency on Aβ1-42 aggregation, with an inhibition value of 74.08% and 78.39% at 100 μM and 55.35% and 61.36% at 25 μM, respectively. Molecular modeling studies were carried out for the most active compound against AChE, compound 2l. All the results suggested that compounds 2d and 2i have better inhibitory potencies on Aβ1-42 aggregation and moderate AChE enzyme activity, and therefore can be highlighted as promising compounds.
Collapse
Affiliation(s)
- Görkem S Fidan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ege University, Izmir, Turkey
| | - Sulunay Parlar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ege University, Izmir, Turkey
| | - Ayse H Tarikogullari
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ege University, Izmir, Turkey
| | - Vildan Alptuzun
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ege University, Izmir, Turkey
| | - Ayşe S Alpan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ege University, Izmir, Turkey
| |
Collapse
|
188
|
Neuroprotective Effects of Phytochemicals against Aluminum Chloride-Induced Alzheimer’s Disease through ApoE4/LRP1, Wnt3/β-Catenin/GSK3β, and TLR4/NLRP3 Pathways with Physical and Mental Activities in a Rat Model. Pharmaceuticals (Basel) 2022; 15:ph15081008. [PMID: 36015156 PMCID: PMC9416484 DOI: 10.3390/ph15081008] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/05/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Alzheimer’s disease (AD) is a neurodegenerative disorder that is associated with abnormal cognition. AD is aided in its initiation and progression by hereditary and environmental factors. Aluminum (Al) is a neurotoxic agent that causes oxidative stress, which is linked to AD progression. Additionally, Nrf2/HO-1, APOE4/LRP1, Wnt3/β-catenin, and TLR4/NLRP3 are the main signaling pathways involved in AD pathogenesis. Several phytochemicals are promising options in delaying AD evolution. Objectives: This study aimed at studying the neuroprotective effects of some phytochemicals as morin (MOR), thymol (TML), and thymoquinone (TMQ) on physical and mental activities (PhM) in Al chloride (AlCl3)-induced AD rat model. Another objective was to determine the specificity of phytochemicals to AD signaling pathways using molecular docking. Methods: Eighty male Dawley rats were divided into eight groups. Each group received: saline (control group), AlCl3, (ALAD), PhM, either alone or with a combination of MOR, TML, and/or TMQ for five weeks. Animals were then subjected to behavioral evaluation. Brain tissues were used for histopathological and biochemical analyses to determine the extent of neurodegeneration. The effect of phytochemicals on AlCl3-induced oxidative stress and the main signaling pathways involved in AD progression were also investigated. Results: AlCl3 caused a decline in spatial learning and memory, as well as histopathological changes in the brains of rats. Phytochemicals combined with PhM restored antioxidant activities, increased HO-1 and Nrf2 levels, blocked inflammasome activation, apoptosis, TLR4 expression, amyloide-β generation, and tau hyperphophorylation. They also brought ApoE4 and LRP1 levels back to normal and regulated Wnt3/β-catenin/GSK3β signaling pathway. Conclusions: The use of phytochemicals with PhM is a promising strategy for reducing AD by modulating Nrf2/HO-1, TLR4/NLRP3, APOE4/LRP1, and Wnt3/β-catenin/GSK-3β signaling pathways.
Collapse
|
189
|
Dhamodharan J, Sekhar G, Muthuraman A. Epidermal Growth Factor Receptor Kinase Inhibitor Ameliorates β-Amyloid Oligomer-Induced Alzheimer Disease in Swiss Albino Mice. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27165182. [PMID: 36014421 PMCID: PMC9412386 DOI: 10.3390/molecules27165182] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/04/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022]
Abstract
Alzheimer's disease (AD) is one of the major neurodegenerative disorders, and its incidence increases globally every year. Currently, available AD drugs symptomatically treat AD with multiple adverse effects. Gefitinib (GE) is an epidermal growth factor receptor (EGFR) kinase inhibitor. EGFR is the preferred target for the treatment of AD, whereas the effect of GE in AD conditions is limited. The present study was designed to explore the ameliorative potential of GE in Aβ1-42 oligomer-induced neurotoxicity in AD mice. AD was induced by intracerebroventricular (i.c.v.) injection of Aβ1-42 oligomer (4 μg/4 μL) into the lateral ventricles of the mouse brain. The test compound, i.e., GE (2 and 4 mg/kg of body weight), was administered orally on days 10, 13, 16, 19, 22, 25, and 28, and the reference drug, i.e., donepezil (DP, 2 mg/kg), was administered orally from the 10th to 28th days. The behavioral changes were screened by the Morris water maze (MWM) test. Furthermore, biomarkers i.e., brain acetylcholinesterase (AChE), thiobarbituric acid reactive substances (TBARS), and reduced glutathione (GSH) levels were estimated from brain samples. The AD-associated histopathological changes were analyzed by hematoxylin and eosin staining. The administration of GE significantly ameliorated the AD-associated behavioral, biochemical, and histopathological changes. The ameliorative effect of GE against the Aβ1-42 oligomer-associated neurotoxicity was due to its potent inhibition of EGFR kinase activation, as well as its antioxidant and antilipid peroxidative effect.
Collapse
Affiliation(s)
- Jagadeesh Dhamodharan
- Unit of Anatomy, Faculty of Medicine, AIMST University, Semeling, Bedong 08100, Kedah, Malaysia
- Department of Pathology, Faculty of Medicine, Saveetha Institute of Medical & Technical Sciences (SIMATS), Saveetha University, Chennai 602105, Tamilnadu, India
| | - Ganthimathy Sekhar
- Department of Pathology, Faculty of Medicine, Saveetha Institute of Medical & Technical Sciences (SIMATS), Saveetha University, Chennai 602105, Tamilnadu, India
| | - Arunachalam Muthuraman
- Unit of Pharmacology, Faculty of Pharmacy, AIMST University, Bedong 08100, Kedah, Malaysia
- Correspondence:
| |
Collapse
|
190
|
Chowdhury MR, Jin HK, Bae JS. Diverse Roles of Ceramide in the Progression and Pathogenesis of Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10081956. [PMID: 36009503 PMCID: PMC9406151 DOI: 10.3390/biomedicines10081956] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/31/2022] [Accepted: 08/02/2022] [Indexed: 11/26/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disorder, and is associated with several pathophysiological features, including cellular dysfunction, failure of neurotransmission, cognitive impairment, cell death, and other clinical consequences. Advanced research on the pathogenesis of AD has elucidated a mechanistic framework and revealed many therapeutic possibilities. Among the mechanisms, sphingolipids are mentioned as distinctive mediators to be associated with the pathology of AD. Reportedly, alteration in the metabolism of sphingolipids and their metabolites result in the dysfunction of mitochondria, autophagy, amyloid beta regulation, and neuronal homeostasis, which exacerbates AD progression. Considering the importance of sphingolipids, in this review, we discuss the role of ceramide, a bioactive sphingolipid metabolite, in the progression and pathogenesis of AD. Herein, we describe the ceramide synthesis pathway and its involvement in the dysregulation of homeostasis, which eventually leads to AD. Furthermore, this review references different therapeutics proposed to modulate the ceramide pathway to maintain ceramide levels and prevent the disease progression.
Collapse
Affiliation(s)
- Md Riad Chowdhury
- KNU Alzheimer’s Disease Research Institute, Kyungpook National University, Daegu 41566, Korea
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Hee Kyung Jin
- KNU Alzheimer’s Disease Research Institute, Kyungpook National University, Daegu 41566, Korea
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea
- Correspondence: (H.K.J.); (J.-s.B.); Tel.: +82-53-950-5966 (H.K.J.); +82-53-420-4815 (J.-s.B.); Fax: +82-53-950-5955 (H.K.J.); +82-53-424-3349 (J.-s.B.)
| | - Jae-sung Bae
- KNU Alzheimer’s Disease Research Institute, Kyungpook National University, Daegu 41566, Korea
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Correspondence: (H.K.J.); (J.-s.B.); Tel.: +82-53-950-5966 (H.K.J.); +82-53-420-4815 (J.-s.B.); Fax: +82-53-950-5955 (H.K.J.); +82-53-424-3349 (J.-s.B.)
| |
Collapse
|
191
|
Guo J, Cheng M, Liu P, Cao D, Luo J, Wan Y, Fang Y, Jin Y, Xie SS, Liu J. A multi-target directed ligands strategy for the treatment of Alzheimer's disease: Dimethyl fumarate plus Tranilast modified Dithiocarbate as AChE inhibitor and Nrf2 activator. Eur J Med Chem 2022; 242:114630. [PMID: 35987018 DOI: 10.1016/j.ejmech.2022.114630] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) possessed intricate pathogenesis. Currently, multi-targeted drugs were considered to have the potential to against AD by simultaneously triggering molecules in functionally complementary pathways. Hence, a series of molecules based on the pharmacophoric features of Dimethyl fumarate, Tranilast, and Dithiocarbate were designed and synthesized. These compounds showed significant AChE inhibitory activity in vitro. Among them, compound 4c2 displayed the mighty inhibitory activity to hAChE (IC50 = 0.053 μM) and held the ability to cross the BBB. Kinetic study and molecular docking pointed out that 4c2 bound well into the active sites of hAChE, forming steady and sturdy interactions with key residues in hAChE. Additionally, 4c2 as an Nrf2 activator could promote the nuclear translocation of Nrf2 protein and induce the expressions of Nrf2-dependent enzymes HO-1, NQO1, and GPX4. Moreover, 4c2 rescued BV-2 cells from H2O2-induced injury and inhibited ROS accumulation. For the anti-neuroinflammatory potential of 4c2, we observed that 4c2 could lower the levels of pro-inflammatory cytokines (NO, IL-6 and TNF-α) and suppressed the expressions of iNOS and COX-2. In particular, 4c2 was well tolerated in mice (2500 mg/kg, p.o.) and efficaciously recovered the memory impairment in a Scopolamine-induced mouse model. Overall, these results highlighted that 4c2 was a promising multi-targeted agent for treating AD.
Collapse
Affiliation(s)
- Jie Guo
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China
| | - Maojun Cheng
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China
| | - Peng Liu
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China
| | - Duanyuan Cao
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China
| | - Jinchong Luo
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China
| | - Yang Wan
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China
| | - Yuanying Fang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China
| | - Yi Jin
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China
| | - Sai-Sai Xie
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China.
| | - Jing Liu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330006, PR China.
| |
Collapse
|
192
|
Multitargeting the Action of 5-HT 6 Serotonin Receptor Ligands by Additional Modulation of Kinases in the Search for a New Therapy for Alzheimer's Disease: Can It Work from a Molecular Point of View? Int J Mol Sci 2022; 23:ijms23158768. [PMID: 35955902 PMCID: PMC9368844 DOI: 10.3390/ijms23158768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/03/2022] [Accepted: 08/05/2022] [Indexed: 11/26/2022] Open
Abstract
In view of the unsatisfactory treatment of cognitive disorders, in particular Alzheimer’s disease (AD), the aim of this review was to perform a computer-aided analysis of the state of the art that will help in the search for innovative polypharmacology-based therapeutic approaches to fight against AD. Apart from 20-year unrenewed cholinesterase- or NMDA-based AD therapy, the hope of effectively treating Alzheimer’s disease has been placed on serotonin 5-HT6 receptor (5-HT6R), due to its proven, both for agonists and antagonists, beneficial procognitive effects in animal models; however, research into this treatment has so far not been successfully translated to human patients. Recent lines of evidence strongly emphasize the role of kinases, in particular microtubule affinity-regulating kinase 4 (MARK4), Rho-associated coiled-coil-containing protein kinase I/II (ROCKI/II) and cyclin-dependent kinase 5 (CDK5) in the etiology of AD, pointing to the therapeutic potential of their inhibitors not only against the symptoms, but also the causes of this disease. Thus, finding a drug that acts simultaneously on both 5-HT6R and one of those kinases will provide a potential breakthrough in AD treatment. The pharmacophore- and docking-based comprehensive literature analysis performed herein serves to answer the question of whether the design of these kind of dual agents is possible, and the conclusions turned out to be highly promising.
Collapse
|
193
|
He Q, Shi L, Luo Y, Wan C, Malone IB, Mok VCT, Cole JH, Anatürk M. Validation of the Alzheimer's disease-resemblance atrophy index in classifying and predicting progression in Alzheimer's disease. Front Aging Neurosci 2022; 14:932125. [PMID: 36062150 PMCID: PMC9435378 DOI: 10.3389/fnagi.2022.932125] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/14/2022] [Indexed: 12/01/2022] Open
Abstract
Background Automated tools for characterising dementia risk have the potential to aid in the diagnosis, prognosis, and treatment of Alzheimer's disease (AD). Here, we examined a novel machine learning-based brain atrophy marker, the AD-resemblance atrophy index (AD-RAI), to assess its test-retest reliability and further validate its use in disease classification and prediction. Methods Age- and sex-matched 44 probable AD (Age: 69.13 ± 7.13; MMSE: 27-30) and 22 non-demented control (Age: 69.38 ± 7.21; MMSE: 27-30) participants were obtained from the Minimal Interval Resonance Imaging in Alzheimer's Disease (MIRIAD) dataset. Serial T1-weighted images (n = 678) from up to nine time points over a 2-year period, including 179 pairs of back-to-back scans acquired on same participants on the same day and 40 pairs of scans acquired at 2-week intervals were included. All images were automatically processed with AccuBrain® to calculate the AD-RAI. Its same-day repeatability and 2-week reproducibility were first assessed. The discriminative performance of AD-RAI was evaluated using the receiver operating characteristic curve, where DeLong's test was used to evaluate its performance against quantitative medial temporal lobe atrophy (QMTA) and hippocampal volume adjusted by intracranial volume (ICV)-proportions and ICV-residuals methods, respectively (HVR and HRV). Linear mixed-effects modelling was used to investigate longitudinal trajectories of AD-RAI and baseline AD-RAI prediction of cognitive decline. Finally, the longitudinal associations between AD-RAI and MMSE scores were assessed. Results AD-RAI had excellent same-day repeatability and excellent 2-week reproducibility. AD-RAI's AUC (99.8%; 95%CI = [99.3%, 100%]) was equivalent to that of QMTA (96.8%; 95%CI = [92.9%, 100%]), and better than that of HVR (86.8%; 95%CI = [78.2%, 95.4%]) or HRV (90.3%; 95%CI = [83.0%, 97.6%]). While baseline AD-RAI was significantly higher in the AD group, it did not show detectable changes over 2 years. Baseline AD-RAI was negatively associated with MMSE scores and the rate of the change in MMSE scores over time. A negative longitudinal association was also found between AD-RAI values and the MMSE scores among AD patients. Conclusions The AD-RAI represents a potential biomarker that may support AD diagnosis and be used to predict the rate of future cognitive decline in AD patients.
Collapse
Affiliation(s)
- Qiling He
- UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom
| | - Lin Shi
- Department of Imaging and Interventional Radiology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Yishan Luo
- BrainNow Research Institute, Shenzhen, China
| | - Chao Wan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ian B. Malone
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Faculty of Brain Sciences, University College London, London, United Kingdom
| | - Vincent C. T. Mok
- Gerald Choa Neuroscience Centre, Therese Pei Fong Chow Research Centre for Prevention of Dementia, Lui Che Woo Institute of Innovative Medicine, Division of Neurology, Department of Medicine and Therapeutics, Prince of Wales Hospital, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - James H. Cole
- Department of Computer Science, Faculty of Engineering Science, University College London, London, United Kingdom
- Dementia Research Centre, Institute of Neurology, University College London, London, United Kingdom
| | - Melis Anatürk
- Department of Computer Science, Faculty of Engineering Science, University College London, London, United Kingdom
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
194
|
Sanjay, Shin JH, Park M, Lee HJ. Cyanidin-3-O-Glucoside Regulates the M1/M2 Polarization of Microglia via PPARγ and Aβ42 Phagocytosis Through TREM2 in an Alzheimer's Disease Model. Mol Neurobiol 2022; 59:5135-5148. [PMID: 35670898 PMCID: PMC9363298 DOI: 10.1007/s12035-022-02873-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 05/02/2022] [Indexed: 12/22/2022]
Abstract
Microglial polarization plays an essential role in the progression and regression of neurodegenerative disorders. Cyanidin-3-O-glucoside (C3G), a dietary anthocyanin found in many fruits and vegetables, has been reported as an antioxidant, anti-inflammatory, and antitumor agent. However, there have been no reports on whether C3G can regulate the M1/M2 shift in an Alzheimer's disease model. We attempted to investigate the effects of C3G on M1/M2 polarization and the mechanism to regulate anti-inflammation and phagocytosis, both in vitro and in vivo. HMC3 cells were treated with β-amyloid (Aβ42) in the presence or absence of 50 μM C3G for different time intervals, and APPswe/PS1ΔE9 mice were orally administered 30 mg/kg/day of C3G for 38 weeks. The in vitro data revealed that C3G could shift the M1 phenotype of microglia to M2 by reducing the expression of M1-specific markers (CD86 and CD80), inflammatory cytokines (IL-Iβ, IL-6, TNF-α), reactive oxygen species, and enhancing the expression of M2-specific markers (CD206 and CD163). The APPswe/PS1ΔE9 mice results were consistent with the in vitro data, indicating a significant reduction in inflammatory cytokines and higher expression of M2-specific markers such as CD206 and Arg1 in C3G-treated Alzheimer's disease model mice. Additionally, C3G was found to upregulate PPARγ expression levels both in vitro and in vivo, whereas a PPARγ antagonist (GW9662) was found to block C3G-mediated effects in vitro. In this study, we confirmed that C3G could regulate microglial polarization by activating PPARγ and eliminating accumulated β-amyloid by enhancing Aβ42 phagocytosis through the upregulation of TREM2.
Collapse
Affiliation(s)
- Sanjay
- Department of Food Science and Biotechnology, College of BioNano Technology, Gachon University, Seongnam-si 13120, Gyeonggi-do, Korea
| | - Jae-Ho Shin
- Department of Biomedical Laboratory Science, Eulji University, Gyeonggi-do 461-713, Seongnam-si, Republic of Korea
| | - Miey Park
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam-si 13120, Gyeonggi-do, Korea.
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam-si 13120, Gyeonggi-do, Korea.
| | - Hae-Jeung Lee
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam-si 13120, Gyeonggi-do, Korea.
- Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam-si 13120, Gyeonggi-do, Korea.
| |
Collapse
|
195
|
Merde İB, Önel GT, Türkmenoğlu B, Gürsoy Ş, Dilek E, Özçelik AB, Uysal M. Synthesis of (
p‐
tolyl)‐3(2
H
)pyridazinone Derivatives as Novel Acetylcholinesterase Inhibitors. ChemistrySelect 2022. [DOI: 10.1002/slct.202201606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- İrem Bozbey Merde
- Department of Pharmaceutical Chemistry Faculty of Pharmacy Erzincan Binali Yıldırım University Yalnızbağ 24002 Erzincan Turkey
| | - Gülce Taşkor Önel
- Department of Analytical Chemistry Faculty of Pharmacy Erzincan Binali Yıldırım University Yalnızbağ 24002 Erzincan Turkey
| | - Burçin Türkmenoğlu
- Department of Analytical Chemistry Faculty of Pharmacy Erzincan Binali Yıldırım University Yalnızbağ 24002 Erzincan Turkey
| | - Şule Gürsoy
- Department of Biochemistry Faculty of Pharmacy Erzincan Binali Yıldırım University Yalnızbağ 24002 Erzincan Turkey
| | - Esra Dilek
- Department of Biochemistry Faculty of Pharmacy Erzincan Binali Yıldırım University Yalnızbağ 24002 Erzincan Turkey
| | - Azime Berna Özçelik
- Department of Pharmaceutical Chemistry Faculty of Pharmacy Gazi University 06330 Ankara Turkey
| | - Mehtap Uysal
- Department of Pharmaceutical Chemistry Faculty of Pharmacy Erzincan Binali Yıldırım University Yalnızbağ 24002 Erzincan Turkey
| |
Collapse
|
196
|
Li M, Cao X, Yan H, Wang M, Tashibolati A, Maiwulanjiang M. Integrating Zebrafish Model to Screen Active Ingredients and Network Pharmacology Methods to Explore the Mechanism of Lavandula angustifolia Therapy for Alzheimer's Disease. ChemistrySelect 2022. [DOI: 10.1002/slct.202201364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Muchun Li
- State Key Laboratory Basis of Xinjiang indigenous medicinal plants resource utilization Xinjiang Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing South Road 40–1 Urumqi 830011 Xinjiang China
- University of Chinese Academy of Sciences Beijing 100049 China
- Xinjiang Academic Institute of Analysis and Testing Plant Resources Green Processing Engineering Technology Research Center of Xinjiang North Science Road 374 Urumqi 830011 Xinjiang China
| | - Xueqin Cao
- State Key Laboratory Basis of Xinjiang indigenous medicinal plants resource utilization Xinjiang Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing South Road 40–1 Urumqi 830011 Xinjiang China
- University of Chinese Academy of Sciences Beijing 100049 China
- Xinjiang Academic Institute of Analysis and Testing Plant Resources Green Processing Engineering Technology Research Center of Xinjiang North Science Road 374 Urumqi 830011 Xinjiang China
| | - Huan Yan
- Xinjiang Academic Institute of Analysis and Testing Plant Resources Green Processing Engineering Technology Research Center of Xinjiang North Science Road 374 Urumqi 830011 Xinjiang China
- College of Public Health Xinjiang Medical University Urumqi 830011 Xinjiang China
| | - Miaomiao Wang
- State Key Laboratory Basis of Xinjiang indigenous medicinal plants resource utilization Xinjiang Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing South Road 40–1 Urumqi 830011 Xinjiang China
- University of Chinese Academy of Sciences Beijing 100049 China
- Xinjiang Academic Institute of Analysis and Testing Plant Resources Green Processing Engineering Technology Research Center of Xinjiang North Science Road 374 Urumqi 830011 Xinjiang China
| | - Ayiguli Tashibolati
- Xinjiang Academic Institute of Analysis and Testing Plant Resources Green Processing Engineering Technology Research Center of Xinjiang North Science Road 374 Urumqi 830011 Xinjiang China
| | - Maitinuer Maiwulanjiang
- State Key Laboratory Basis of Xinjiang indigenous medicinal plants resource utilization Xinjiang Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing South Road 40–1 Urumqi 830011 Xinjiang China
| |
Collapse
|
197
|
Choi HJ, Park JH, Jeong YJ, Hwang JW, Lee S, Lee H, Seol E, Kim IW, Cha BY, Seo J, Moon M, Hoe HS. Donepezil ameliorates Aβ pathology but not tau pathology in 5xFAD mice. Mol Brain 2022; 15:63. [PMID: 35850693 PMCID: PMC9290238 DOI: 10.1186/s13041-022-00948-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/06/2022] [Indexed: 11/15/2022] Open
Abstract
The cholinesterase inhibitor donepezil is used to improve Aβ pathology and cognitive function in patients with Alzheimer’s disease (AD). However, the impact of donepezil on tau pathology is unclear. Thus, we examined the effects of donepezil on Aβ and tau pathology in 5xFAD mice (a model of AD) in this study. We found that intraperitoneal injection of donepezil (1 mg/kg, i.p.) exhibited significant reductions in Aβ plaque number in the cortex and hippocampal DG region. In addition, donepezil treatment (1 mg/kg, i.p.) reduced Aβ-mediated microglial and, to a lesser extent, astrocytic activation in 5xFAD mice. However, neither intraperitoneal/oral injection of donepezil nor oral injection of rivastigmine altered tau phosphorylation at Thr212/Ser214 (AT100), Thr396, and Thr231 in 5xFAD mice. Surprisingly, we observed that intraperitoneal/oral injection of donepezil treatment significantly increased tau phosphorylation at Thr212 in 5xFAD mice. Taken together, these data suggest that intraperitoneal injection of donepezil suppresses Aβ pathology but not tau pathology in 5xFAD mice.
Collapse
Affiliation(s)
- Hee-Jeong Choi
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, 41068, Daegu, Republic of Korea
| | - Jin-Hee Park
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, 41068, Daegu, Republic of Korea.,Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, 42988, Daegu, Korea
| | - Yoo Joo Jeong
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, 41068, Daegu, Republic of Korea
| | - Jeong-Woo Hwang
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, 41068, Daegu, Republic of Korea
| | - Soojung Lee
- G2GBIO, Inc., Science Park#411, 1646 Yuseond-daero, Yuseong-gu, 34054, Daejeon, Korea
| | - Heeyong Lee
- G2GBIO, Inc., Science Park#411, 1646 Yuseond-daero, Yuseong-gu, 34054, Daejeon, Korea
| | - Eunyoung Seol
- G2GBIO, Inc., Science Park#411, 1646 Yuseond-daero, Yuseong-gu, 34054, Daejeon, Korea
| | - Ik-Whi Kim
- PharmacoRex Co., Ltd., 20 Techno 1-ro, Yuseong-gu, 34016, Daejeon, Korea
| | - Byung-Yoon Cha
- PharmacoRex Co., Ltd., 20 Techno 1-ro, Yuseong-gu, 34016, Daejeon, Korea
| | - Jinsoo Seo
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, 42988, Daegu, Korea
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, 35365, Daejeon, Korea.
| | - Hyang-Sook Hoe
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, 41068, Daegu, Republic of Korea. .,Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, 42988, Daegu, Korea.
| |
Collapse
|
198
|
Wang P, Yu L, Gong J, Xiong J, Zi S, Xie H, Zhang F, Mao Z, Liu Z, Kim JS. An Activity‐Based Fluorescent Probe for Imaging Fluctuations of Peroxynitrite (ONOO‐) in the Alzheimer's Disease Brain. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202206894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Pengzhan Wang
- Ministry of education key laboratory for the synthesis and application of organic functional molecules, Hubei University, Wuhan College of chemistry and chemical engineering 430062 CHINA
| | - Le Yu
- Korea university, Seoul Chemistry KOREA, REPUBLIC OF
| | - Jiankang Gong
- Ministry of education key laboratory for the synthesis and application of organic functional molecules College of chemistry and chemical engineering 430062 CHINA
| | - Jianhua Xiong
- Wuhan university, Wuhan College of chemistry and molecular science CHINA
| | - Soyu Zi
- Korea university, Seoul Chemistry KOREA, REPUBLIC OF
| | - Hua Xie
- Wuhan University, Wuhan School of water resources and hydropower CHINA
| | - Fan Zhang
- Ministry of educational key laboratory for the synthesis and application of organic functional molecules, Hubei University, Wuhan College of chemistry and chemical engineering CHINA
| | - Zhiqiang Mao
- Ministry of education key laboratory for the synthesis and application of organic functional molecules, Huibei University, Wuhan College of chemistry and chemical engineering CHINA
| | - Zhihong Liu
- Ministry of education key laboratory for the synthesis and application of organic functional molecules, Huibei University, Wuhan College of chemistry and chemical engineering CHINA
| | - Jong Seung Kim
- Korea University Department of Chemistry Anamdong 02841 Seoul KOREA, REPUBLIC OF
| |
Collapse
|
199
|
Faldu KG, Shah JS. Alzheimer's disease: a scoping review of biomarker research and development for effective disease diagnosis. Expert Rev Mol Diagn 2022; 22:681-703. [PMID: 35855631 DOI: 10.1080/14737159.2022.2104639] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 07/19/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is regarded as the foremost reason for neurodegeneration that prominently affects the geriatric population. Characterized by extracellular accumulation of amyloid-beta (Aβ), intracellular aggregation of hyperphosphorylated tau (p-tau), and neuronal degeneration that causes impairment of memory and cognition. Amyloid/tau/neurodegeneration (ATN) classification is utilized for research purposes and involves amyloid, tau, and neuronal injury staging through MRI, PET scanning, and CSF protein concentration estimations. CSF sampling is invasive, and MRI and PET scanning requires sophisticated radiological facilities which limit its widespread diagnostic use. ATN classification lacks effectiveness in preclinical AD. AREAS COVERED This publication intends to collate and review the existing biomarker profile and the current research and development of a new arsenal of biomarkers for AD pathology from different biological samples, microRNA (miRNA), proteomics, metabolomics, artificial intelligence, and machine learning for AD screening, diagnosis, prognosis, and monitoring of AD treatments. EXPERT OPINION It is an accepted observation that AD-related pathological changes occur over a long period of time before the first symptoms are observed providing ample opportunity for detection of biological alterations in various biological samples that can aid in early diagnosis and modify treatment outcomes.
Collapse
Affiliation(s)
- Khushboo Govind Faldu
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Jigna Samir Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India
| |
Collapse
|
200
|
Roles of Fatty Acids in Microglial Polarization: Evidence from In Vitro and In Vivo Studies on Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms23137300. [PMID: 35806302 PMCID: PMC9266841 DOI: 10.3390/ijms23137300] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023] Open
Abstract
Microglial polarization to the M1 phenotype (classically activated) or the M2 phenotype (alternatively activated) is critical in determining the fate of immune responses in neurodegenerative diseases (NDs). M1 macrophages contribute to neurotoxicity, neuronal and synaptic damage, and oxidative stress and are the first line of defense, and M2 macrophages elicit an anti-inflammatory response to regulate neuroinflammation, clear cell debris, and promote neuroregeneration. Various studies have focused on the ability of natural compounds to promote microglial polarization from the M1 phenotype to the M2 phenotype in several diseases, including NDs. However, studies on the roles of fatty acids in microglial polarization and their implications in NDs are a rare find. Most of the studies support the role of polyunsaturated fatty acids (PUFAs) in microglial polarization using cell and animal models. Thus, we aimed to collect data and provide a narrative account of microglial types, markers, and studies pertaining to fatty acids, particularly PUFAs, on microglial polarization and their neuroprotective effects. The involvement of only PUFAs in the chosen topic necessitates more in-depth research into the role of unexplored fatty acids in microglial polarization and their mechanistic implications. The review also highlights limitations and future challenges.
Collapse
|