251
|
Sanderson-Cimino M, Panizzon MS, Elman JA, Tu X, Gustavson DE, Puckett O, Cross K, Notestine R, Hatton SN, Eyler LT, McEvoy LK, Hagler DJ, Neale MC, Gillespie NA, Lyons MJ, Franz CE, Fennema-Notestine C, Kremen WS. Periventricular and deep abnormal white matter differ in associations with cognitive performance at midlife. Neuropsychology 2021; 35:252-264. [PMID: 33970659 PMCID: PMC8500190 DOI: 10.1037/neu0000718] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Objective: Abnormal white matter (AWM) on magnetic resonance imaging is associated with cognitive performance in older adults. We explored cognitive associations with AWM during late-midlife. Method: Participants were community-dwelling men (n = 242; M = 61.90 years; range = 56-66). Linear-mixed effects regression models examined associations of total, periventricular, and deep AWM with cognitive performance, controlling for multiple comparisons. Models considering specific cognitive domains controlled for current general cognitive ability (GCA). We hypothesized that total AWM would be associated with worse processing speed, executive function, and current GCA; deep AWM would correlate with GCA and periventricular AWM would relate to specific cognitive abilities. We also assessed the potential influence of cognitive reserve by examining a moderation effect of early life (mean age of 20) cognition. Results: Greater total and deep AWM were associated with poorer current GCA. Periventricular AWM was associated with worse executive function, working memory, and episodic memory. When periventricular and deep AWM were modeled simultaneously, both retained their respective significant associations with cognitive performance. Cognitive reserve did not moderate associations. Conclusions: Our findings suggest that AWM contributes to poorer cognitive function in late-midlife. Examining only total AWM may obscure the potential differential impact of regional AWM. Separating total AWM into subtypes while controlling for current GCA revealed a dissociation in relationships with cognitive performance; deep AWM was associated with nonspecific cognitive ability whereas periventricular AWM was associated with specific frontal-related abilities and memory. Management of vascular or other risk factors that may increase the risk of AWM should begin during or before early late-midlife. (PsycInfo Database Record (c) 2021 APA, all rights reserved).
Collapse
Affiliation(s)
- Mark Sanderson-Cimino
- Joint Doctoral Program in Clinical Psychology, San Diego State/University of California
- Center for Behavior Genetics of Aging, University of California
| | - Matthew S. Panizzon
- Center for Behavior Genetics of Aging, University of California
- Department of Psychiatry University of California
| | - Jeremy A. Elman
- Center for Behavior Genetics of Aging, University of California
- Department of Psychiatry University of California
| | - Xin Tu
- Family Medicine and Public Health, University of California
| | - Daniel E. Gustavson
- Center for Behavior Genetics of Aging, University of California
- Department of Psychiatry University of California
- Department of Medicine, Vanderbilt University Medical Center
| | - Olivia Puckett
- Center for Behavior Genetics of Aging, University of California
- Department of Psychiatry University of California
| | | | - Randy Notestine
- Department of Psychiatry University of California
- Computational and Applied Statistics Laboratory (CASL) at the San Diego Supercomputer Center
| | - Sean N Hatton
- Center for Behavior Genetics of Aging, University of California
- Department of Psychiatry University of California
- Department of Neurosciences, University of California
| | - Lisa T. Eyler
- Department of Psychiatry University of California
- Mental Illness Research, Education, And Clinical Center, Veterans Affairs San Diego Healthcare System
| | - Linda K. McEvoy
- Department of Radiology, University of California, San Diego
| | | | - Michael C. Neale
- Virginia Institute for Psychiatric and Behavior Genetics, Virginia Commonwealth University
| | - Nathan A. Gillespie
- Virginia Institute for Psychiatric and Behavior Genetics, Virginia Commonwealth University
| | - Michael J. Lyons
- Department of Psychological and Brain Sciences, Boston University
| | - Carol E. Franz
- Center for Behavior Genetics of Aging, University of California
- Department of Psychiatry University of California
| | - Christine Fennema-Notestine
- Center for Behavior Genetics of Aging, University of California
- Department of Psychiatry University of California
- Department of Radiology, University of California, San Diego
| | - William S. Kremen
- Center for Behavior Genetics of Aging, University of California
- Department of Psychiatry University of California
- Center of Excellence for Stress and Mental Health, Veterans Affairs San Diego Healthcare System
| |
Collapse
|
252
|
Sonawane SK, Uversky VN, Chinnathambi S. Baicalein inhibits heparin-induced Tau aggregation by initializing non-toxic Tau oligomer formation. Cell Commun Signal 2021; 19:16. [PMID: 33579328 PMCID: PMC7879681 DOI: 10.1186/s12964-021-00704-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 01/02/2021] [Indexed: 12/14/2022] Open
Abstract
Background Amyloid aggregate deposition is the key feature of Alzheimer’s disease. The proteinaceous aggregates found in the afflicted brain are the intra-neuronal neurofibrillary tangles formed by the microtubule-associated protein Tau and extracellular deposits, senile plaques, of amyloid beta (Aβ) peptide proteolytically derived from the amyloid precursor protein. Accumulation of these aggregates has manifestations in the later stages of the disease, such as memory loss and cognitive inabilities originating from the neuronal dysfunction, neurodegeneration, and brain atrophy. Treatment of this disease at the late stages is difficult, and many clinical trials have failed. Hence, the goal is to find means capable of preventing the aggregation of these intrinsically disordered proteins by inhibiting the early stages of their pathological transformations. Polyphenols are known to be neuroprotective agents with the noticeable potential against many neurodegenerative diseases, such as Alzheimer’s, Parkinson’s, and Prion diseases. Methods We analyzed the capability of Baicalein to inhibit aggregation of human Tau protein by a multifactorial analysis that included several biophysical and biochemical techniques. Results The potency of Baicalein, a polyphenol from the Scutellaria baicalensis Georgi, against in vitro Tau aggregation and PHF dissolution has been screened and validated. ThS fluorescence assay revealed the potent inhibitory activity of Baicalein, whereas ANS revealed its mechanism of Tau inhibition viz. by oligomer capture and dissociation. In addition, Baicalein dissolved the preformed mature fibrils of Tau thereby possessing a dual target action. Tau oligomers formed by Baicalein were non-toxic to neuronal cells, highlighting its role as a potent molecule to be screened against AD. Conclusion In conclusion, Baicalein inhibits aggregation of hTau40 by enhancing the formation of SDS-stable oligomers and preventing fibril formation. Baicalein-induced oligomers do not affect the viability of the neuroblastoma cells. Therefore, Baicalein can be considered as a lead molecule against Tau pathology in AD. Video Abstract
Collapse
Affiliation(s)
- Shweta Kishor Sonawane
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory (CSIR-NCL), Dr. Homi Bhabha Road, Pune, 411008, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Subashchandrabose Chinnathambi
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory (CSIR-NCL), Dr. Homi Bhabha Road, Pune, 411008, India. .,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
253
|
CD33 rs3865444 as a risk factor for Parkinson's disease. Neurosci Lett 2021; 748:135709. [PMID: 33582190 DOI: 10.1016/j.neulet.2021.135709] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/29/2021] [Accepted: 01/31/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Alzheimer's (AD) and Parkinson's diseases (PD) share a few elements of their clinical, pathological and genetic backgrounds. The CD33 rs3865444 has emerged as a strong genetic locus associated with AD through genome-wide association study (GWAS). However, little is known for its role in PD. OBJECTIVE To assess the role of CD33 rs3865444 on PD risk. METHODS We genotyped 358 patients with PD and 358 healthy controls for theCD33 rs3865444. Odds ratios (ORs) with the respective 95% confidence intervals (CIs)], were calculated with the SNPStats software, assuming five genetic models (co-dominant, dominant, recessive, over-dominant and log-additive), with the G allele as the reference allele. RESULTS The CD33 rs3865444 was associated with decreased PD risk in the dominant [GG vs GT + TT; OR (95% CI) = 0.61 (0.45-0.82), p = 0.001], the over-dominant [GG + TT vs GT; OR (95% CI) = 0.65 (0.48-0.89), p = 0.0061], log-additive [OR (95% CI) = 0.67 (0.52-0.86), p = 0.0014], and co-dominant [with overall p = 0.0043, and OR (95% CI) = 0.62 (0.45-0.84) for the TG genotype compared to the GG], modes of inheritance. CONCLUSIONS The CD33 rs3865444 is associated with decreased PD risk, and larger studies investigating the role of CD33 rs3865444 on PD are needed.
Collapse
|
254
|
Paek EJ, Yoon SO. Partner-Specific Communication Deficits in Individuals With Alzheimer's Disease. AMERICAN JOURNAL OF SPEECH-LANGUAGE PATHOLOGY 2021; 30:376-390. [PMID: 32585126 DOI: 10.1044/2020_ajslp-19-00094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Purpose Speakers adjust referential expressions to the listeners' knowledge while communicating, a phenomenon called "audience design." While individuals with Alzheimer's disease (AD) show difficulties in discourse production, it is unclear whether they exhibit preserved partner-specific audience design. The current study examined if individuals with AD demonstrate partner-specific audience design skills. Method Ten adults with mild-to-moderate AD and 12 healthy older adults performed a referential communication task with two experimenters (E1 and E2). At first, E1 and participants completed an image-sorting task, allowing them to establish shared labels. Then, during testing, both experimenters were present in the room, and participants described images to either E1 or E2 (randomly alternating). Analyses focused on the number of words participants used to describe each image and whether they reused shared labels. Results During testing, participants in both groups produced shorter descriptions when describing familiar images versus new images, demonstrating their ability to learn novel knowledge. When they described familiar images, healthy older adults modified their expressions depending on the current partner's knowledge, producing shorter expressions and more established labels for the knowledgeable partner (E1) versus the naïve partner (E2), but individuals with AD were less likely to do so. Conclusions The current study revealed that both individuals with AD and the control participants were able to acquire novel knowledge, but individuals with AD tended not to flexibly adjust expressions depending on the partner's knowledge state. Conversational inefficiency and difficulties observed in AD may, in part, stem from disrupted audience design skills.
Collapse
Affiliation(s)
- Eun Jin Paek
- Department of Audiology and Speech Pathology, The University of Tennessee Health Science Center, Knoxville
| | - Si On Yoon
- Department of Communication Sciences and Disorders, The University of Iowa, Iowa City
| |
Collapse
|
255
|
Potential of Naturally Derived Alkaloids as Multi-Targeted Therapeutic Agents for Neurodegenerative Diseases. Molecules 2021; 26:molecules26030728. [PMID: 33573300 PMCID: PMC7866829 DOI: 10.3390/molecules26030728] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 01/26/2021] [Accepted: 01/26/2021] [Indexed: 01/11/2023] Open
Abstract
Alkaloids are a class of secondary metabolites that can be derived from plants, fungi and marine sponges. They are widely known as a continuous source of medicine for the management of chronic disease including cancer, diabetes and neurodegenerative diseases. For example, galanthamine and huperzine A are alkaloid derivatives currently being used for the symptomatic management of neurodegenerative disease. The etiology of neurodegenerative diseases is polygenic and multifactorial including but not limited to inflammation, oxidative stress and protein aggregation. Therefore, natural-product-based alkaloids with polypharmacology modulation properties are potentially useful for further drug development or, to a lesser extent, as nutraceuticals to manage neurodegeneration. This review aims to discuss and summarise recent developments in relation to naturally derived alkaloids for neurodegenerative diseases.
Collapse
|
256
|
Rossi A, Galla L, Gomiero C, Zentilin L, Giacca M, Giorgio V, Calì T, Pozzan T, Greotti E, Pizzo P. Calcium Signaling and Mitochondrial Function in Presenilin 2 Knock-Out Mice: Looking for Any Loss-of-Function Phenotype Related to Alzheimer's Disease. Cells 2021; 10:204. [PMID: 33494218 PMCID: PMC7909802 DOI: 10.3390/cells10020204] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/17/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is the most common age-related neurodegenerative disorder in which learning, memory and cognitive functions decline progressively. Familial forms of AD (FAD) are caused by mutations in amyloid precursor protein (APP), presenilin 1 (PSEN1) and presenilin 2 (PSEN2) genes. Presenilin 1 (PS1) and its homologue, presenilin 2 (PS2), represent, alternatively, the catalytic core of the γ-secretase complex that, by cleaving APP, produces neurotoxic amyloid beta (Aβ) peptides responsible for one of the histopathological hallmarks in AD brains, the amyloid plaques. Recently, PSEN1 FAD mutations have been associated with a loss-of-function phenotype. To investigate whether this finding can also be extended to PSEN2 FAD mutations, we studied two processes known to be modulated by PS2 and altered by FAD mutations: Ca2+ signaling and mitochondrial function. By exploiting neurons derived from a PSEN2 knock-out (PS2-/-) mouse model, we found that, upon IP3-generating stimulation, cytosolic Ca2+ handling is not altered, compared to wild-type cells, while mitochondrial Ca2+ uptake is strongly compromised. Accordingly, PS2-/- neurons show a marked reduction in endoplasmic reticulum-mitochondria apposition and a slight alteration in mitochondrial respiration, whereas mitochondrial membrane potential, and organelle morphology and number appear unchanged. Thus, although some alterations in mitochondrial function appear to be shared between PS2-/- and FAD-PS2-expressing neurons, the mechanisms leading to these defects are quite distinct between the two models. Taken together, our data appear to be difficult to reconcile with the proposal that FAD-PS2 mutants are loss-of-function, whereas the concept that PS2 plays a key role in sustaining mitochondrial function is here confirmed.
Collapse
Affiliation(s)
- Alice Rossi
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (A.R.); (L.G.); (C.G.); (V.G.); (T.C.); (T.P.); (P.P.)
| | - Luisa Galla
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (A.R.); (L.G.); (C.G.); (V.G.); (T.C.); (T.P.); (P.P.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
| | - Chiara Gomiero
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (A.R.); (L.G.); (C.G.); (V.G.); (T.C.); (T.P.); (P.P.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
| | - Lorena Zentilin
- International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy; (L.Z.); (M.G.)
| | - Mauro Giacca
- International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy; (L.Z.); (M.G.)
| | - Valentina Giorgio
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (A.R.); (L.G.); (C.G.); (V.G.); (T.C.); (T.P.); (P.P.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
- Department of Biomedical and Neuromotor Science, University of Bologna, 40112 Bologna, Italy
| | - Tito Calì
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (A.R.); (L.G.); (C.G.); (V.G.); (T.C.); (T.P.); (P.P.)
| | - Tullio Pozzan
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (A.R.); (L.G.); (C.G.); (V.G.); (T.C.); (T.P.); (P.P.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
- Venetian Institute of Molecular Medicine (VIMM), 35131 Padua, Italy
| | - Elisa Greotti
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (A.R.); (L.G.); (C.G.); (V.G.); (T.C.); (T.P.); (P.P.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
| | - Paola Pizzo
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (A.R.); (L.G.); (C.G.); (V.G.); (T.C.); (T.P.); (P.P.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
| |
Collapse
|
257
|
Zhang N, Zhao L, Su Y, Liu X, Zhang F, Gao Y. Syringin Prevents Aβ 25-35-Induced Neurotoxicity in SK-N-SH and SK-N-BE Cells by Modulating miR-124-3p/BID Pathway. Neurochem Res 2021; 46:675-685. [PMID: 33471295 DOI: 10.1007/s11064-021-03240-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 11/24/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder disease, disturbing people's normal life. Syringin was mentioned to antagonize Amyloid-β (Aβ)-induced neurotoxicity. However, the action mechanism is still not fully elucidated. This study aimed to explore a molecular mechanism of syringin in defending Aβ-induced neurotoxicity. SK-N-SH and SK-N-BE cells were treated with amyloid β-protein fragment 25-35 (Aβ25-35) to induce cell neurotoxicity. The injury effects were distinguished by assessing cell viability and cell apoptosis using cell counting kit-8 (CCK-8) assay and flow cytometry assay, respectively. The expression of Cleaved-caspase3 (Cleaved-casp3), B cell lymphoma/leukemia-2 (Bcl-2), Bcl-2 associated X protein (Bax) and BH3 interacting domain death agonist (BID) at the protein level was determined by western blot. The expression of miR-124-3p and BID was detected using quantitative real-time polymerase chain reaction (qRT-PCR). The interaction between miR-124-3p and BID was predicted by the online database starBase and confirmed by dual-luciferase reporter assay plus RNA pull-down assay. Aβ25-35 treatment inhibited cell viability and induced cell apoptosis, while the addition of syringin recovered cell viability and suppressed cell apoptosis. MiR-124-3p was significantly downregulated in Aβ25-35-treated SK-N-SH and SK-N-BE cells, and BID was upregulated. Nevertheless, the addition of syringin reversed their expression. BID was a target of miR-124-3p, and its downregulation partly prevented Aβ25-35-induced injuries. Syringin protected against Aβ25-35-induced neurotoxicity by enhancing miR-124-3p expression and weakening BID expression, and syringin strengthened the expression of miR-124-3p to diminish BID level. Syringin ameliorated Aβ25-35-induced neurotoxicity in SK-N-SH and SK-N-BE cells by regulating miR-124-3p/BID pathway, which could be a novel theoretical basis for syringin to treat AD.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Geriatrics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Li Zhao
- Department of Pharmacy, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Yan Su
- Medical Service, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Xiaoliang Liu
- Department of Neurosurgery, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Feilong Zhang
- Public Health Division, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Yiwen Gao
- Department of Pharmacy, Yantai Affiliated Hospital of Binzhou Medical University, No. 717, Jinbu Street, MuPing District, Yantai, 264100, Shandong, China.
| |
Collapse
|
258
|
Farugia TL, Cuni-Lopez C, White AR. Potential Impacts of Extreme Heat and Bushfires on Dementia. J Alzheimers Dis 2021; 79:969-978. [PMID: 33459654 DOI: 10.3233/jad-201388] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Australia often experiences natural disasters and extreme weather conditions such as: flooding, sandstorms, heatwaves, and bushfires (also known as wildfires or forest fires). The proportion of the Australian population aged 65 years and over is increasing, alongside the severity and frequency of extreme weather conditions and natural disasters. Extreme heat can affect the entire population but particularly at the extremes of life, and patients with morbidities. Frequently identified as a vulnerable demographic in natural disasters, there is limited research on older adults and their capacity to deal with extreme heat and bushfires. There is a considerable amount of literature that suggests a significant association between mental disorders such as dementia, and increased vulnerability to extreme heat. The prevalence rate for dementia is estimated at 30%by age 85 years, but there has been limited research on the effects extreme heat and bushfires have on individuals living with dementia. This review explores the differential diagnosis of dementia, the Australian climate, and the potential impact Australia's extreme heat and bushfires have on individuals from vulnerable communities including low socioeconomic status Indigenous and Non-Indigenous populations living with dementia, in both metropolitan and rural communities. Furthermore, we investigate possible prevention strategies and provide suggestions for future research on the topic of Australian bushfires and heatwaves and their impact on people living with dementia. This paper includes recommendations to ensure rural communities have access to appropriate support services, medical treatment, awareness, and information surrounding dementia.
Collapse
Affiliation(s)
- Taya L Farugia
- Mental Health Program, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Carla Cuni-Lopez
- Mental Health Program, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Anthony R White
- Mental Health Program, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| |
Collapse
|
259
|
Neuroprotective Effects of 2-Substituted 1, 3-Selenazole Amide Derivatives on Amyloid-Beta-Induced Toxicity in a Transgenic Caenorhabditis Elegans Model of Alzheimer's Disease. Neurotox Res 2021; 39:841-850. [PMID: 33400180 DOI: 10.1007/s12640-020-00321-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease is an age-related neurodegenerative disease, associated with the presence of extracellular amyloid-β (Aβ) plaques and neurofibrillary tangles. Although the pathogenesis of AD remains unclear, the characteristic feature of AD was reported to be the buildup of Aβ plaques. In this study, we extensively investigated the neuroprotective effects of 2-substituted 1,3-selenazole amide derivatives (CHF11) on Aβ1-42 transgenic Caenorhabditis elegans CL4176. Results showed that worms fed with CHF11 exhibited remarkably reduced paralysis, decreased levels of toxic Aβ oligomers and Aβ plaque deposition, as well as less ROS production in comparison with the untreated worms. The effective concentrations of CHF11 were arranged in the descending order of 100 µM > 10 µM > 1 µM. Real-time PCR analysis showed that there was no significant difference in Aβ expression between CHF11-administered group and the blank control group, suggesting that CHF11-induced reduction in toxic protein deposition may be regulated at the post-transcriptional level. In the meantime, the gene expressions of hsf-1 and its downstream target hsp-12.6 were significantly increased, indicating that CHF11 against Aβ toxicity may involve in HSF-1 signaling pathway in worms. In conclusion, CHF11 exhibits a significant protective effect against β-amyloid-induced toxicity in CL4176 by reducing β-amyloid aggregation and ROS production, which may involve in HSF-1 and downstream target HSP-12.6 pathway.
Collapse
|
260
|
Platycodin D Inhibits β-Amyloid-Induced Inflammation and Oxidative Stress in BV-2 Cells Via Suppressing TLR4/NF-κB Signaling Pathway and Activating Nrf2/HO-1 Signaling Pathway. Neurochem Res 2021; 46:638-647. [PMID: 33394221 DOI: 10.1007/s11064-020-03198-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 10/22/2022]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease associated with deposition of β-amyloid peptide (Aβ). Platycodin D (PLD), a triterpenesaponin, may possess neuro-protective effect. In the current study, we aimed to explore the effects of PLD on Aβ-induced inflammation and oxidative stress in microglial BV-2 cells. Our study showed that PLD treatment improved cell viability in Aβ-induced BV-2 cells. PLD attenuated Aβ-induced inflammation with deceased production of TNF-α, IL-1β and IL-6 in Aβ-induced BV-2 cells. PLD also mitigated the oxidative stress in Aβ-induced BV-2 cells, as evidenced by deceased production of ROS and MDA, and increased SOD activity. Furthermore, the increased expression levels of TLR4 and p-p65 and decreased IκBα expression in the Aβ-stimulated BV-2 cells were attenuated by PLD treatment. Overexpression of TLR4 reversed the anti-inflammatory effect of PLD in Aβ-stimulated BV-2 cells. In addition, PLD treatment enhanced the Aβ-stimulated increase in the expression levels of Nrf2, HO-1, and NQO1 in BV-2 cells. Knockdown of Nrf2 abrogated the anti-oxidative effect of PLD in Aβ-stimulated BV-2 cells. In conclusion, these findings indicated that PLD protected BV-2 cells from Aβ-induced oxidative stress and inflammation via regulating the TLR4/NF-κB and Nrf2/HO-1 signaling pathways. Thus, PLD may be a potential candidate for the treatment of AD.
Collapse
|
261
|
Shekari A, Fahnestock M. Cholinergic neurodegeneration in Alzheimer disease mouse models. HANDBOOK OF CLINICAL NEUROLOGY 2021; 182:191-209. [PMID: 34266592 DOI: 10.1016/b978-0-12-819973-2.00013-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Cholinergic signaling is critical for cognitive function. The basal forebrain is the major cholinergic output of the central nervous system. Degeneration of basal forebrain cholinergic neurons is a hallmark of Alzheimer's disease (AD). Mouse models are invaluable tools in disease research and have been used to study AD for over 25 years. However, animal models of AD vary greatly with respect to the degree of cholinergic degeneration observed. The following review will outline the most influential animal models of AD with an emphasis on the basal forebrain cholinergic system.
Collapse
Affiliation(s)
- Arman Shekari
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Margaret Fahnestock
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
262
|
Panchal K, Tiwari AK. Miro (Mitochondrial Rho GTPase), a key player of mitochondrial axonal transport and mitochondrial dynamics in neurodegenerative diseases. Mitochondrion 2021; 56:118-135. [PMID: 33127590 DOI: 10.1016/j.mito.2020.10.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/14/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023]
Abstract
Miro (mitochondrial Rho GTPases) a mitochondrial outer membrane protein, plays a vital role in the microtubule-based mitochondrial axonal transport, mitochondrial dynamics (fusion and fission) and Mito-Ca2+ homeostasis. It forms a major protein complex with Milton (an adaptor protein), kinesin and dynein (motor proteins), and facilitates bidirectional mitochondrial axonal transport such as anterograde and retrograde transport. By forming this protein complex, Miro facilitates the mitochondrial axonal transport and fulfills the neuronal energy demand, maintain the mitochondrial homeostasis and neuronal survival. It has been demonstrated that altered mitochondrial biogenesis, improper mitochondrial axonal transport, and mitochondrial dynamics are the early pathologies associated with most of the neurodegenerative diseases (NDs). Being the sole mitochondrial outer membrane protein associated with mitochondrial axonal transport-related processes, Miro proteins can be one of the key players in various NDs such as Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic lateral sclerosis (ALS) and Huntington's disease (HD). Thus, in the current review, we have discussed the evolutionarily conserved Miro proteins and its role in the pathogenesis of the various NDs. From this, we indicated that Miro proteins may act as a potential target for a novel therapeutic intervention for the treatment of various NDs.
Collapse
Affiliation(s)
- Komal Panchal
- Genetics & Developmental Biology Laboratory, Department of Biological Sciences & Biotechnology, Institute of Advanced Research (IAR), Koba, Gandhinagar, Gujarat 382426, India
| | - Anand Krishna Tiwari
- Genetics & Developmental Biology Laboratory, Department of Biological Sciences & Biotechnology, Institute of Advanced Research (IAR), Koba, Gandhinagar, Gujarat 382426, India.
| |
Collapse
|
263
|
Xie J, Zi W, Li Z, He Y. Ontology-based Precision Vaccinology for Deep Mechanism Understanding and Precision Vaccine Development. Curr Pharm Des 2021; 27:900-910. [PMID: 33238868 PMCID: PMC12067353 DOI: 10.2174/1381612826666201125112131] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 10/08/2020] [Indexed: 11/22/2022]
Abstract
Vaccination is one of the most important innovations in human history. It has also become a hot research area in a new application - the development of new vaccines against non-infectious diseases such as cancers. However, effective and safe vaccines still do not exist for many diseases, and where vaccines exist, their protective immune mechanisms are often unclear. Although licensed vaccines are generally safe, various adverse events, and sometimes severe adverse events, still exist for a small population. Precision medicine tailors medical intervention to the personal characteristics of individual patients or sub-populations of individuals with similar immunity-related characteristics. Precision vaccinology is a new strategy that applies precision medicine to the development, administration, and post-administration analysis of vaccines. Several conditions contribute to make this the right time to embark on the development of precision vaccinology. First, the increased level of research in vaccinology has generated voluminous "big data" repositories of vaccinology data. Secondly, new technologies such as multi-omics and immunoinformatics bring new methods for investigating vaccines and immunology. Finally, the advent of AI and machine learning software now makes possible the marriage of Big Data to the development of new vaccines in ways not possible before. However, something is missing in this marriage, and that is a common language that facilitates the correlation, analysis, and reporting nomenclature for the field of vaccinology. Solving this bioinformatics problem is the domain of applied biomedical ontology. Ontology in the informatics field is human- and machine-interpretable representation of entities and the relations among entities in a specific domain. The Vaccine Ontology (VO) and Ontology of Vaccine Adverse Events (OVAE) have been developed to support the standard representation of vaccines, vaccine components, vaccinations, host responses, and vaccine adverse events. Many other biomedical ontologies have also been developed and can be applied in vaccine research. Here, we review the current status of precision vaccinology and how ontological development will enhance this field, and propose an ontology-based precision vaccinology strategy to support precision vaccine research and development.
Collapse
Affiliation(s)
- Jiangan Xie
- Chongqing Engineering Research Center of Medical Electronics and Information Technology, School of Bioinformatics, Chongqing University of Posts and Telecommunications, Chongqing, China
| | - Wenrui Zi
- Chongqing Engineering Research Center of Medical Electronics and Information Technology, School of Bioinformatics, Chongqing University of Posts and Telecommunications, Chongqing, China
| | - Zhangyong Li
- Chongqing Engineering Research Center of Medical Electronics and Information Technology, School of Bioinformatics, Chongqing University of Posts and Telecommunications, Chongqing, China
| | - Yongqun He
- Unit of Laboratory Animal Medicine, Development of Microbiology and Immunology, Center of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
264
|
Jiskoot LC, Poos JM, Vollebergh ME, Franzen S, van Hemmen J, Papma JM, van Swieten JC, Kessels RPC, van den Berg E. Emotion recognition of morphed facial expressions in presymptomatic and symptomatic frontotemporal dementia, and Alzheimer's dementia. J Neurol 2021; 268:102-113. [PMID: 32728945 PMCID: PMC7815624 DOI: 10.1007/s00415-020-10096-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 07/10/2020] [Accepted: 07/20/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND The emotion recognition task (ERT) was developed to overcome shortcomings of static emotion recognition paradigms, by identifying more subtle deficits in emotion recognition across different intensity levels. In this study, we used the ERT to investigate emotion recognition deficits across the frontotemporal (FTD) and Alzheimer's Dementia (AD) spectrum. METHODS With the ERT, we assessed the recognition of facial emotional expressions (anger-disgust-fear-happiness-sadness-surprise) across four intensities (40-60-80-100%) in patients with behavioural variant FTD (bvFTD; n = 32), and AD (n = 32), presymptomatic FTD mutation carriers (n = 47) and controls (n = 49). We examined group differences using multilevel linear regression with age, sex and education level as covariates, and performed post hoc analyses on presymptomatic (MAPT, GRN and C9orf72) mutation carriers. Classification abilities were investigated by means of logistic regression. RESULTS Lowest ERT total scores were found in patients with bvFTD and AD, whereas equal highest performance was found in presymptomatic mutation carriers and controls. For all emotions, significantly lower subscores were found in patients with bvFTD than in presymptomatic mutation carriers and in controls (highest p value = 0.025). Patients with bvFTD performed lower than patients with AD on anger (p = 0.005) and a trend towards significance was found for a lower performance on happiness (p = 0.065). Task performance increased with higher emotional intensity, and classification was better at the lowest than at the highest intensity. C9orf72 mutation carriers performed worse on recognizing anger at the lowest intensity than GRN mutation carriers (p = 0.047) and controls (p = 0.038). The ERT differentiated between patients with bvFTD and controls, and between patients with AD and controls (both p < 0.001). DISCUSSION Our results demonstrate emotion recognition deficits in both bvFTD and AD, and suggest the presence of subtle emotion recognition changes in presymptomatic C9orf72-FTD. This highlights the importance of incorporating emotion recognition paradigms into standard neuropsychological assessment for early differential diagnosis, and as clinical endpoints in upcoming therapeutic trials.
Collapse
Affiliation(s)
- Lize C. Jiskoot
- Department of Neurology, Erasmus Medical Center, NF-331, Post box 2040, 3000 CA Rotterdam, The Netherlands
- Dementia Research Centre, University College London, London, UK
| | - Jackie M. Poos
- Department of Neurology, Erasmus Medical Center, NF-331, Post box 2040, 3000 CA Rotterdam, The Netherlands
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Manon E. Vollebergh
- Department of Neurology, Erasmus Medical Center, NF-331, Post box 2040, 3000 CA Rotterdam, The Netherlands
| | - Sanne Franzen
- Department of Neurology, Erasmus Medical Center, NF-331, Post box 2040, 3000 CA Rotterdam, The Netherlands
| | - Judy van Hemmen
- Department of Neurology, Erasmus Medical Center, NF-331, Post box 2040, 3000 CA Rotterdam, The Netherlands
| | - Janne M. Papma
- Department of Neurology, Erasmus Medical Center, NF-331, Post box 2040, 3000 CA Rotterdam, The Netherlands
| | - John C. van Swieten
- Department of Neurology, Erasmus Medical Center, NF-331, Post box 2040, 3000 CA Rotterdam, The Netherlands
| | - Roy P. C. Kessels
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, The Netherlands
- Department of Medical Psychology, Radboudumc Alzheimer Center, Nijmegen, The Netherlands
| | - Esther van den Berg
- Department of Neurology, Erasmus Medical Center, NF-331, Post box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
265
|
Sahu B, Mackos AR, Floden AM, Wold LE, Combs CK. Particulate Matter Exposure Exacerbates Amyloid-β Plaque Deposition and Gliosis in APP/PS1 Mice. J Alzheimers Dis 2021; 80:761-774. [PMID: 33554902 PMCID: PMC8100996 DOI: 10.3233/jad-200919] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by the accumulation of amyloid-β (Aβ) plaques, neuroinflammation, and neuronal death. There are several well-established genetic and environmental factors hypothesized to contribute to AD progression including air pollution. However, the molecular mechanisms by which air pollution exacerbates AD are unclear. OBJECTIVE This study explored the effects of particulate matter exposure on AD-related brain changes using the APP/PS1 transgenic model of disease. METHODS Male C57BL/6;C3H wild type and APP/PS1 mice were exposed to either filtered air (FA) or particulate matter sized under 2.5μm (PM2.5) for 6 h/day, 5 days/week for 3 months and brains were collected. Immunohistochemistry for Aβ, GFAP, Iba1, and CD68 and western blot analysis for PS1, BACE, APP, GFAP, and Iba1 were performed. Aβ ELISAs and cytokine arrays were performed on frozen hippocampal and cortical lysates, respectively. RESULTS The Aβ plaque load was significantly increased in the hippocampus of PM2.5-exposed APP/PS1 mice compared to their respective FA controls. Additionally, in the PM2.5-exposed APP/PS1 group, increased astrocytosis and microgliosis were observed as indicated by elevated GFAP, Iba1, and CD68 immunoreactivities. PM2.5 exposure also led to an elevation in the levels of PS1 and BACE in APP/PS1 mice. The cytokines TNF-α, IL-6, IL-1β, IFN-γ, and MIP-3α were also elevated in the cortices of PM2.5-exposed APP/PS1 mice compared to FA controls. CONCLUSION Our data suggest that chronic particulate matter exposure exacerbates AD by increasing Aβ plaque load, gliosis, and the brain inflammatory status.
Collapse
Affiliation(s)
- Bijayani Sahu
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND 58202-9037
| | - Amy R. Mackos
- College of Nursing, The Ohio State University, Columbus, OH
| | - Angela M. Floden
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND 58202-9037
| | - Loren E. Wold
- College of Nursing, The Ohio State University, Columbus, OH
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH
| | - Colin K. Combs
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, 1301 N Columbia Road, Grand Forks, ND 58202-9037
| |
Collapse
|
266
|
Ahmed AS. JAK-1/STAT-3 pathway mediated role in aging cerebellar cortex degenerative changes of albino wistar rats. TRANSLATIONAL RESEARCH IN ANATOMY 2021. [DOI: 10.1016/j.tria.2020.100089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
267
|
Mroczek M, Desouky A, Sirry W. Imaging Transcriptomics in Neurodegenerative Diseases. J Neuroimaging 2020; 31:244-250. [PMID: 33368775 DOI: 10.1111/jon.12827] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/24/2020] [Accepted: 12/03/2020] [Indexed: 11/30/2022] Open
Abstract
Imaging transcriptomics investigates the relationship between neuroanatomical/neuroimaging features and gene expression. The spatial and temporal distribution of the expressed genes and their pattern of spreading over time can contribute to elucidating cellular and molecular processes involved in neurodegeneration. In this study, we review recent findings regarding the correlation between neuroimaging and expression data in neurodegenerative diseases with a focus on Alzheimer's disease and Parkinson's disease. An association between gene expression data and different neuroimaging neurodegeneration features, such as R2 relaxation time and volumetric cortical atrophy, was established. Several positive and negative expression correlations were identified, and they confirmed the focal nature of neurodegeneration. Positively correlated genes were associated with cell motility, immune system activity, neuroinflammation, and microglia. Data from connectome studies support the hypothesis of selective network vulnerability and a temporal spreading pattern in neurodegenerative pathologies. Genes related to cellular mobility and transport are overexpressed in the neuroimaging-defined delineated areas of degeneration. In addition, expression enrichment of genes involved in immunological processes in vulnerable regions-such as the Toll-like receptor, a receptor involved in innate immunity-plays a major role in neuroinflammation in neurodegenerative diseases. However, substantial limitations must be overcome in future studies: the lack of high-quality resolution expression data, the lack of standardized study protocols, and insufficient sensitive early stage neuroimaging markers of degeneration. Identifying neuroimaging and expression prodromal biomarkers and investigating their causal relation in the preclinical disease stage may enable early targeted therapy before the onset of irreversible brain changes.
Collapse
Affiliation(s)
- Magdalena Mroczek
- Centre for Gerontopsychiatric Medicine, Department of Geriatric Psychiatry, University Hospital of Psychiatry Zürich, Zürich, Switzerland
| | - Ahmed Desouky
- School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Wadid Sirry
- Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
268
|
Agarwal M, Alam MR, Haider MK, Malik MZ, Kim DK. Alzheimer's Disease: An Overview of Major Hypotheses and Therapeutic Options in Nanotechnology. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 11:E59. [PMID: 33383712 PMCID: PMC7823376 DOI: 10.3390/nano11010059] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/18/2020] [Accepted: 12/24/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD), a progressively fatal neurodegenerative disorder, is the most prominent form of dementia found today. Patients suffering from Alzheimer's begin to show the signs and symptoms, like decline in memory and cognition, long after the cellular damage has been initiated in their brain. There are several hypothesis for the neurodegeneration process; however, the lack of availability of in vivo models makes the recapitulation of AD in humans impossible. Moreover, the drugs currently available in the market serve to alleviate the symptoms and there is no cure for the disease. There have been two major hurdles in the process of finding the same-the inefficiency in cracking the complexity of the disease pathogenesis and the inefficiency in delivery of drugs targeted for AD. This review discusses the different drugs that have been designed over the recent years and the drug delivery options in the field of nanotechnology that have been found most feasible in surpassing the blood-brain barrier (BBB) and reaching the brain.
Collapse
Affiliation(s)
- Mugdha Agarwal
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida 201309, India;
| | - Mohammad Rizwan Alam
- Department of Medical Genetics, School of Medicine, Keimyung University, Daegu 42601, Korea;
| | | | - Md. Zubbair Malik
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Dae-Kwang Kim
- Department of Medical Genetics, School of Medicine, Keimyung University, Daegu 42601, Korea;
- Hanvit Institute for Medical Genetics, Daegu 42601, Korea
| |
Collapse
|
269
|
Arroyo-Anlló EM, Souchaud C, Ingrand P, Chamorro Sánchez J, Melero Ventola A, Gil R. Alexithymia in Alzheimer's Disease. J Clin Med 2020; 10:jcm10010044. [PMID: 33375608 PMCID: PMC7795069 DOI: 10.3390/jcm10010044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 11/16/2022] Open
Abstract
Alexithymia is widely recognized as the inability to identify and express emotions. It is a construct which consists of four cognitive traits such as difficulty in identifying feelings, describing feelings to others, externally oriented thinking, and limited imaginative capacity. Several studies have linked alexithymia to cognitive functioning, observing greater alexithymia scores associated with poorer cognitive abilities. Despite Alzheimer's disease (AD) being a neurodegenerative pathology characterized by cognitive troubles from the early stages, associated to behavioral and emotional disturbances, very few investigations have studied the alexithymia in AD. These studies have shown that alexithymia scores-assessed with Toronto Alexithymia Scale (TAS)-were greater in AD patients than healthy participants. The objective of the study was to investigate if the alexithymia was present in patients with mild AD. We hypothesized that the AD group would show more alexithymia features than the control group. We evaluated 54 subjects, including 27 patients diagnosed with mild AD and 27 normal healthy controls, using the Shalling Sifneos Psychosomatic Scale (SSPS-R) and a neuropsychological test battery. Using non-parametric statistical analyses-Wilcoxon and Mann-Whitney U tests-we observed that the SSPS-R scores were similar in the AD and control groups. All participants showed SSPS-R scores below to 10 points, which means no-alexithymia. We did not find significant correlations between SSPS-R scores and cognitive variables in both groups (p > 0.22), but we observed a negative association between name abilities and alexithymia, but it does not reach to significance (p = 0.07). However, a significant correlation between SSPS-R score and mood state, assessed using Zerssen Rating Scale, was found in both groups (p = 0.01). Because we did not find a significant difference in the alexithymia assessment between both subject groups, pot hoc analyses were computed for each item of the SSPS-R. We made comparisons of alexithymic responses percentages in each SSPS-R item between AD and control groups, using Fisher's test. We observed that AD patients produced more alexithymic responses in some items of SSPS-R test than the control group, particularly about difficulties to find the words to describe feelings, as well as difficulties of imagination capacity and externally oriented thinking. The present results do not confirm our hypothesis and they do not support the results of previous studies revealing great alexithymia in AD.
Collapse
Affiliation(s)
- Eva Mª Arroyo-Anlló
- Department of Psychobiology, Neuroscience Institute of Castilla-León, University of Salamanca, 37007 Salamanca, Spain
- Correspondence: ; Tel.: +34-629460944
| | - Corinne Souchaud
- Department of Neurology and Neuropsychology, University Hospital, CHU La Milétrie, 86000 Poitiers, France;
| | - Pierre Ingrand
- Department of Biostatistics, University of Poitiers, 86000 Poitiers, France;
| | - Jorge Chamorro Sánchez
- Faculty of Psychology, Pontifical University of Salamanca, 37002 Salamanca, Spain; (J.C.S.); (A.M.V.)
| | - Alejandra Melero Ventola
- Faculty of Psychology, Pontifical University of Salamanca, 37002 Salamanca, Spain; (J.C.S.); (A.M.V.)
| | - Roger Gil
- Department of Neurology, University Hospital, 86000 Poitiers, France;
| |
Collapse
|
270
|
Konecny J, Misiachna A, Hrabinova M, Pulkrabkova L, Benkova M, Prchal L, Kucera T, Kobrlova T, Finger V, Kolcheva M, Kortus S, Jun D, Valko M, Horak M, Soukup O, Korabecny J. Pursuing the Complexity of Alzheimer's Disease: Discovery of Fluoren-9-Amines as Selective Butyrylcholinesterase Inhibitors and N-Methyl-d-Aspartate Receptor Antagonists. Biomolecules 2020; 11:biom11010003. [PMID: 33375115 PMCID: PMC7822176 DOI: 10.3390/biom11010003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/14/2020] [Accepted: 12/18/2020] [Indexed: 12/13/2022] Open
Abstract
Alzheimer’s disease (AD) is a complex disorder with unknown etiology. Currently, only symptomatic therapy of AD is available, comprising cholinesterase inhibitors and N-methyl-d-aspartate (NMDA) receptor antagonists. Drugs targeting only one pathological condition have generated only limited efficacy. Thus, combining two or more therapeutic interventions into one molecule is believed to provide higher benefit for the treatment of AD. In the presented study, we designed, synthesized, and biologically evaluated 15 novel fluoren-9-amine derivatives. The in silico prediction suggested both the oral availability and permeation through the blood–brain barrier (BBB). An initial assessment of the biological profile included determination of the cholinesterase inhibition and NMDA receptor antagonism at the GluN1/GluN2A and GluN1/GluN2B subunits, along with a low cytotoxicity profile in the CHO-K1 cell line. Interestingly, compounds revealed a selective butyrylcholinesterase (BChE) inhibition pattern with antagonistic activity on the NMDARs. Their interaction with butyrylcholinesterase was elucidated by studying enzyme kinetics for compound 3c in tandem with the in silico docking simulation. The docking study showed the interaction of the tricyclic core of new derivatives with Trp82 within the anionic site of the enzyme in a similar way as the template drug tacrine. From the kinetic analysis, it is apparent that 3c is a competitive inhibitor of BChE.
Collapse
Affiliation(s)
- Jan Konecny
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic; (J.K.); (M.H.); (L.P.); (T.K.); (D.J.)
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; (M.B.); (L.P.); (T.K.); (V.F.)
| | - Anna Misiachna
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic; (A.M.); (M.K.); (S.K.); (M.H.)
- Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
- Department of Physiology, Faculty of Science, Charles University in Prague, Albertov 6, 128 43 Prague, Czech Republic
| | - Martina Hrabinova
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic; (J.K.); (M.H.); (L.P.); (T.K.); (D.J.)
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; (M.B.); (L.P.); (T.K.); (V.F.)
| | - Lenka Pulkrabkova
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic; (J.K.); (M.H.); (L.P.); (T.K.); (D.J.)
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; (M.B.); (L.P.); (T.K.); (V.F.)
| | - Marketa Benkova
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; (M.B.); (L.P.); (T.K.); (V.F.)
| | - Lukas Prchal
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; (M.B.); (L.P.); (T.K.); (V.F.)
| | - Tomas Kucera
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic; (J.K.); (M.H.); (L.P.); (T.K.); (D.J.)
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; (M.B.); (L.P.); (T.K.); (V.F.)
| | - Tereza Kobrlova
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; (M.B.); (L.P.); (T.K.); (V.F.)
| | - Vladimir Finger
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; (M.B.); (L.P.); (T.K.); (V.F.)
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic
| | - Marharyta Kolcheva
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic; (A.M.); (M.K.); (S.K.); (M.H.)
- Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Stepan Kortus
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic; (A.M.); (M.K.); (S.K.); (M.H.)
- Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Daniel Jun
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic; (J.K.); (M.H.); (L.P.); (T.K.); (D.J.)
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; (M.B.); (L.P.); (T.K.); (V.F.)
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, Radlinskeho 9, 812 37 Bratislava, Slovakia;
| | - Martin Horak
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic; (A.M.); (M.K.); (S.K.); (M.H.)
- Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Ondrej Soukup
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic; (J.K.); (M.H.); (L.P.); (T.K.); (D.J.)
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; (M.B.); (L.P.); (T.K.); (V.F.)
- Correspondence: (O.S.); (J.K.); Tel.: +420-495-833-447 (O.S. & J.K.)
| | - Jan Korabecny
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic; (J.K.); (M.H.); (L.P.); (T.K.); (D.J.)
- Biomedical Research Centre, University Hospital Hradec Kralove, Sokolska 581, 500 05 Hradec Kralove, Czech Republic; (M.B.); (L.P.); (T.K.); (V.F.)
- Correspondence: (O.S.); (J.K.); Tel.: +420-495-833-447 (O.S. & J.K.)
| |
Collapse
|
271
|
TuĞrak M, GÜl Hİ, Anil B, GÜlÇİn İ. Synthesis and pharmacological effects of novel benzenesulfonamides carrying benzamide moiety as carbonic anhydrase and acetylcholinesterase inhibitors. Turk J Chem 2020; 44:1601-1609. [PMID: 33488256 PMCID: PMC7763114 DOI: 10.3906/kim-2007-37] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/08/2020] [Indexed: 01/24/2023] Open
Abstract
N
-(1-(4-Methoxyphenyl)-3-oxo-3-((4-(
N
-(substituted)sulfamoyl)phenyl)amino)prop-1-en-1-yl)benzamides
3a – g
were designed since sulfonamide and benzamide pharmacophores draw great attention in novel drug design due to their wide range of bioactivities including acetylcholinesterase (AChE) and human carbonic anhydrase I and II (hCA I and hCA II) inhibitory potencies. Structure elucidation of the compounds was carried out by 1H NMR, 13C NMR, and HRMS spectra. In vitro enzyme assays showed that the compounds had significant inhibitory potential against hCA I, hCA II, and AChE enzymes at nanomolar levels. Ki values were in the range of 4.07 ± 0.38 – 29.70 ± 3.18 nM for hCA I and 10.68 ± 0.98 – 37.16 ± 7.55 nM for hCA II while Ki values for AChE were in the range of 8.91 ± 1.65 – 34.02 ± 5.90 nM. The most potent inhibitors
3g
(Ki = 4.07 ± 0.38 nM, hCA I),
3c
(Ki = 10.68 ± 0.98 nM, hCA II
)
, and
3f
(Ki = 8.91 ± 1.65 nM, AChE) can be considered as lead compounds of this study with their promising bioactivity results. Secondary sulfonamides showed promising enzyme inhibitory effects on AChE while primary sulfonamide derivative was generally effective on hCA I and hCA II isoenzymes.
Collapse
Affiliation(s)
- Mehtap TuĞrak
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Atatürk University, Erzurum Turkey
| | - Halise İnci GÜl
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Atatürk University, Erzurum Turkey
| | - Barış Anil
- Department of Chemistry, Faculty of Science, Atatürk University, Erzurum Turkey
| | - İlhami GÜlÇİn
- Department of Chemistry, Faculty of Science, Atatürk University, Erzurum Turkey
| |
Collapse
|
272
|
Armstrong TD, Suwannasual U, Kennedy CL, Thasma A, Schneider LJ, Phillippi D, Lund AK. Exposure to Traffic-Generated Pollutants Exacerbates the Expression of Factors Associated with the Pathophysiology of Alzheimer’s Disease in Aged C57BL/6 Wild-Type Mice. J Alzheimers Dis 2020; 78:1453-1471. [DOI: 10.3233/jad-200929] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background: Multiple studies report a strong correlation between traffic-generated air pollution-exposure and detrimental outcomes in the central nervous system (CNS), including Alzheimer’s disease (AD). Incidence of AD is rapidly increasing and, worldwide, many live in regions where pollutants exceed regulatory standards. Thus, it is imperative to identify environmental pollutants that contribute to AD, and the mechanisms involved. Objective: We investigated the effects of mixed gasoline and diesel engine emissions (MVE) on the expression of factors involved in progression of AD in the hippocampus and cerebrum in a young versus aged mouse model. Methods: Young (2 months old) and aged (18 months old) male C57BL/6 mice were exposed to either MVE (300μg/m3 PM) or filtered air (FA) for 6 h/d, 7 d/wk, for 50 d. Immunofluorescence and RT-qPCR were used to quantify oxidative stress (8-OHdG) and expression of amyloid-β protein precursor (AβPP), β secretase (BACE1), amyloid-β (Aβ), aryl hydrocarbon receptor (AhR), cytochrome P450 (CYP) 1B1, angiotensin-converting enzyme (ACE1), and angiotensin II type 1 (AT1) receptor in the cerebrum and hippocampus, in addition to cerebral microvascular tight junction (TJ) protein expression. Results: We observed age-related increases in oxidative stress, AhR, CYP1B1, Aβ, BACE1, and AT1 receptor in the CA1 region of the hippocampus, and elevation of cerebral AβPP, AhR, and CYP1B1 mRNA, associated with decreased cerebral microvascular TJ protein claudin-5. MVE-exposure resulted in further promotion of oxidative stress, and significant increases in AhR, CYP1B1, BACE1, ACE1, and Aβ, compared to the young and aged FA-exposed mice. Conclusion: Such findings suggest that MVE-exposure exacerbates the expression of factors in the CNS associated with AD pathogenesis in aged populations.
Collapse
Affiliation(s)
- Tyler D. Armstrong
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Usa Suwannasual
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Conner L. Kennedy
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Akshaykumar Thasma
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Leah J. Schneider
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Danielle Phillippi
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Amie K. Lund
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| |
Collapse
|
273
|
Pozueta A, Lage C, García-Martínez M, Kazimierczak M, Bravo M, López-García S, Riancho J, González-Suarez A, Vázquez-Higuera JL, de Arcocha-Torres M, Banzo I, Jiménez-Bonilla J, Berciano J, Rodríguez-Rodríguez E, Sánchez-Juan P. Cognitive and Behavioral Profiles of Left and Right Semantic Dementia: Differential Diagnosis with Behavioral Variant Frontotemporal Dementia and Alzheimer's Disease. J Alzheimers Dis 2020; 72:1129-1144. [PMID: 31683488 DOI: 10.3233/jad-190877] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Semantic dementia (SD) is a subtype of frontotemporal dementia (FTD) characterized by semantic memory loss and preserved abilities of other cognitive functions. The clinical manifestations of SD require a differential diagnosis with Alzheimer's disease (AD), especially those with early onset, and behavioral variant FTD (bvFTD). OBJECTIVE The present study aimed to compare cognitive performances and neuropsychiatric symptoms in a population of AD, bvFTD, and left and right SD defined with the support of molecular imaging (amyloid and 2-[18F] fluoro-2-deoxy-D-glucose positron emission tomography) and assessed the accuracy of different neuropsychological markers in distinguishing these neurodegenerative diseases. METHODS Eighty-seven participants (32 AD, 20 bvFTD, and 35 SD (17 Left-SD and 18 Right-SD) completed a comprehensive neuropsychological battery that included memory, language, attention and executive functions, visuospatial function, visuoconstructional skills, and tasks designed specifically to evaluate prosopagnosia and facial emotions recognition. The Neuropsychiatric Inventory was administered to assess neuropsychiatric symptoms. RESULTS An episodic memory test that included semantic cues, a visuospatial test (both impaired in AD), a naming test and a prosopagnosia task (both impaired in SD) were the four most valuable cognitive metrics for the differential diagnosis between groups. Several behavioral abnormalities were differentially present, of which aggression, self-care (both more frequent in bvFTD), and eating habits, specifically overeating and altered dietary preference (more frequent in SD), were the most valuable in group discrimination. CONCLUSION Our study highlights the value of a comprehensive neuropsychological and neuropsychiatric evaluation for the differential diagnosis between FTD syndromes and AD.
Collapse
Affiliation(s)
- Ana Pozueta
- Neurology Service and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 'Marqués de Valdecilla' University Hospital, University of Cantabria, Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
| | - Carmen Lage
- Neurology Service and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 'Marqués de Valdecilla' University Hospital, University of Cantabria, Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
| | - María García-Martínez
- Neurology Service and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 'Marqués de Valdecilla' University Hospital, University of Cantabria, Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
| | - Martha Kazimierczak
- Neurology Service and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 'Marqués de Valdecilla' University Hospital, University of Cantabria, Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
| | - María Bravo
- Neurology Service and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 'Marqués de Valdecilla' University Hospital, University of Cantabria, Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
| | - Sara López-García
- Neurology Service and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 'Marqués de Valdecilla' University Hospital, University of Cantabria, Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
| | - Javier Riancho
- Neurology Service and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Hospital Sierrallana, Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
| | - Andrea González-Suarez
- Neurology Service and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 'Marqués de Valdecilla' University Hospital, University of Cantabria, Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
| | - José Luis Vázquez-Higuera
- Neurology Service and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 'Marqués de Valdecilla' University Hospital, University of Cantabria, Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
| | - María de Arcocha-Torres
- Department of Nuclear Medicine, University Hospital 'Marqués de Valdecilla', University of Cantabria, Molecular imaging Group - IDIVAL, Santander, Spain
| | - Ignacio Banzo
- Department of Nuclear Medicine, University Hospital 'Marqués de Valdecilla', University of Cantabria, Molecular imaging Group - IDIVAL, Santander, Spain
| | - Julio Jiménez-Bonilla
- Department of Nuclear Medicine, University Hospital 'Marqués de Valdecilla', University of Cantabria, Molecular imaging Group - IDIVAL, Santander, Spain
| | - José Berciano
- Neurology Service and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 'Marqués de Valdecilla' University Hospital, University of Cantabria, Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
| | - Eloy Rodríguez-Rodríguez
- Neurology Service and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 'Marqués de Valdecilla' University Hospital, University of Cantabria, Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
| | - Pascual Sánchez-Juan
- Neurology Service and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 'Marqués de Valdecilla' University Hospital, University of Cantabria, Institute for Research 'Marqués de Valdecilla' (IDIVAL), Santander, Spain
| |
Collapse
|
274
|
Reiter K, Gustaw Rothenberg K. Neuropsychological presentation of colpocephaly and porencephaly with symptom onset in adulthood. Neurocase 2020; 26:353-359. [PMID: 33136527 DOI: 10.1080/13554794.2020.1841798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Colpocephaly is a form of congenital ventriculomegaly while porencephaly describes any full-thickness defect within the brain which usually presents as a cystic structure. Postulated aetologies include intrauterine/perinatal injuries, genetic disorders, and morphogenesis error. Colopocephaly and porencephaly is typically diagnosed in infancy while diagnosis in adulthood is exceptionally rare. We report a case of co-existence of colpocephaly with porencephaly diagnosed incidentally in a 54-year-old male presenting with subtle cognitive and neurologic abnormalities. Neuropsychological assessment revealed weaknesses in executive functions, processing speed, and language.To our knowledge, this is the only reported case of dual incidental findings of porencephaly and colpocephaly in an adult.
Collapse
|
275
|
Sadighbayan D, Hasanzadeh M, Ghafar-Zadeh E. Biosensing based on field-effect transistors (FET): Recent progress and challenges. Trends Analyt Chem 2020; 133:116067. [PMID: 33052154 PMCID: PMC7545218 DOI: 10.1016/j.trac.2020.116067] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The use of field-Effect-Transistor (FET) type biosensing arrangements has been highlighted by researchers in the field of early biomarker detection and drug screening. Their non-metalized gate dielectrics that are exposed to an electrolyte solution cover the semiconductor material and actively transduce the biological changes on the surface. The efficiency of these novel devices in detecting different biomolecular analytes in a real-time, highly precise, specific, and label-free manner has been validated by numerous research studies. Considerable progress has been attained in designing FET devices, especially for biomedical diagnosis and cell-based assays in the past few decades. The exceptional electronic properties, compactness, and scalability of these novel tools are very desirable for designing rapid, label-free, and mass detection of biomolecules. With the incorporation of nanotechnology, the performance of biosensors based on FET boosts significantly, particularly, employment of nanomaterials such as graphene, metal nanoparticles, single and multi-walled carbon nanotubes, nanorods, and nanowires. Besides, their commercial availability, and high-quality production on a large-scale, turn them to be one of the most preferred sensing and screening platforms. This review presents the basic structural setup and working principle of different types of FET devices. We also focused on the latest progression regarding the use of FET biosensors for the recognition of viruses such as, recently emerged COVID-19, Influenza, Hepatitis B Virus, protein biomarkers, nucleic acids, bacteria, cells, and various ions. Additionally, an outline of the development of FET sensors for investigations related to drug development and the cellular investigation is also presented. Some technical strategies for enhancing the sensitivity and selectivity of detection in these devices are addressed as well. However, there are still certain challenges which are remained unaddressed concerning the performance and clinical use of transistor-based point-of-care (POC) instruments; accordingly, expectations about their future improvement for biosensing and cellular studies are argued at the end of this review.
Collapse
Affiliation(s)
- Deniz Sadighbayan
- Biologically Inspired Sensors and Actuators (BioSA), Faculty of Science, Dept. of Biology, York University, Toronto, Canada
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hasanzadeh
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ebrahim Ghafar-Zadeh
- Biologically Inspired Sensors and Actuators (BioSA), Faculty of Science, Dept. of Biology, York University, Toronto, Canada
- Dept. of Elecrical Engineering and Computer Science, Lassonde School of Engineering, York University, Toronto, Canada
| |
Collapse
|
276
|
Altamirano‐Espino JA, Sánchez‐Labastida LA, Martínez‐Archundia M, Andrade‐Jorge E, Trujillo‐Ferrara JG. Acetylcholinesterase Inhibition (Potential Anti‐Alzheimer Effects) by Aminobenzoic Acid Derivatives: Synthesis, in Vitro and in Silico Evaluation. ChemistrySelect 2020. [DOI: 10.1002/slct.202003471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- José A. Altamirano‐Espino
- Laboratorio de Investigación en Bioquímica Sección de Estudios de Posgrado e Investigación Escuela Superior de Medicina del Instituto Politécnico Nacional Plan de San Luis y Díaz Mirón s/n Casco de Santo Tomás 11340 Mexico City México
| | - Luis A. Sánchez‐Labastida
- Laboratorio de Investigación en Bioquímica Sección de Estudios de Posgrado e Investigación Escuela Superior de Medicina del Instituto Politécnico Nacional Plan de San Luis y Díaz Mirón s/n Casco de Santo Tomás 11340 Mexico City México
| | - Marlet Martínez‐Archundia
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotécnológica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation) Escuela Superior de Medicina del Instituto Politécnico Nacional Plan de San Luis y Díaz Mirón s/n Casco de Santo Tomás 11340 Mexico City México
| | - Erik Andrade‐Jorge
- Laboratorio de Investigación en Bioquímica Sección de Estudios de Posgrado e Investigación Escuela Superior de Medicina del Instituto Politécnico Nacional Plan de San Luis y Díaz Mirón s/n Casco de Santo Tomás 11340 Mexico City México
- Unidad de Investigación en Biomedicina y Carrera de enfermería Facultad de Estudios Superiores-Iztacala Universidad Nacional Autónoma de México Av. de los Barrios 1, Los Reyes Iztacala Tlalnepantla 54090 Estado de México México
| | - José G. Trujillo‐Ferrara
- Laboratorio de Investigación en Bioquímica Sección de Estudios de Posgrado e Investigación Escuela Superior de Medicina del Instituto Politécnico Nacional Plan de San Luis y Díaz Mirón s/n Casco de Santo Tomás 11340 Mexico City México
| |
Collapse
|
277
|
Martínez-Viturro CM, Trabanco AA, Royes J, Fernández E, Tresadern G, Vega JA, del Cerro A, Delgado F, García Molina A, Tovar F, Shaffer P, Ebneth A, Bretteville A, Mertens L, Somers M, Alonso JM, Bartolomé-Nebreda JM. Diazaspirononane Nonsaccharide Inhibitors of O-GlcNAcase (OGA) for the Treatment of Neurodegenerative Disorders. J Med Chem 2020; 63:14017-14044. [DOI: 10.1021/acs.jmedchem.0c01479] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Carlos M. Martínez-Viturro
- Discovery Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75A, 45007 Toledo, Spain
| | - Andrés A. Trabanco
- Discovery Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75A, 45007 Toledo, Spain
| | - Jordi Royes
- Department Química Física i Inorgànica, University Rovira i Virgili, C/Marcel·lí Domingo s/n, 43007 Tarragona, Spain
| | - Elena Fernández
- Department Química Física i Inorgànica, University Rovira i Virgili, C/Marcel·lí Domingo s/n, 43007 Tarragona, Spain
| | - Gary Tresadern
- Computational Chemistry, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2440 Beerse, Belgium
| | - Juan A. Vega
- Discovery Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75A, 45007 Toledo, Spain
| | - Alcira del Cerro
- Discovery Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75A, 45007 Toledo, Spain
| | - Francisca Delgado
- Discovery Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75A, 45007 Toledo, Spain
| | - Aránzazu García Molina
- Discovery Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75A, 45007 Toledo, Spain
| | - Fulgencio Tovar
- Villapharma Research S.L., Parque Tecnológico de Fuente Álamo, Ctra. El Estrecho-Lobosillo, Km. 2.5—Av. Azul, 30320 Fuente Álamo de Murcia, Spain
| | - Paul Shaffer
- X-Ray Crystallography, Janssen Pharmaceutical Research & Development, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Andreas Ebneth
- Neuroscience Discovery, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2440 Beerse, Belgium
| | - Alexis Bretteville
- Neuroscience Discovery, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2440 Beerse, Belgium
| | - Liesbeth Mertens
- Neuroscience Discovery, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2440 Beerse, Belgium
| | - Marijke Somers
- Discovery DMPK, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2440 Beerse, Belgium
| | - Jose M. Alonso
- Analytical Sciences, Janssen Research & Development, Janssen-Cilag, S.A., C/Jarama 75A, 45007 Toledo, Spain
| | - José M. Bartolomé-Nebreda
- Discovery Chemistry, Janssen Research & Development, Janssen-Cilag S.A., C/Jarama 75A, 45007 Toledo, Spain
| |
Collapse
|
278
|
Spanos F, Liddelow SA. An Overview of Astrocyte Responses in Genetically Induced Alzheimer's Disease Mouse Models. Cells 2020; 9:E2415. [PMID: 33158189 PMCID: PMC7694249 DOI: 10.3390/cells9112415] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/26/2020] [Accepted: 11/02/2020] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. Despite many years of intense research, there is currently still no effective treatment. Multiple cell types contribute to disease pathogenesis, with an increasing body of data pointing to the active participation of astrocytes. Astrocytes play a pivotal role in the physiology and metabolic functions of neurons and other cells in the central nervous system. Because of their interactions with other cell types, astrocyte functions must be understood in their biologic context, thus many studies have used mouse models, of which there are over 190 available for AD research. However, none appear able to fully recapitulate the many functional changes in astrocytes reported in human AD brains. Our review summarizes the observations of astrocyte biology noted in mouse models of familial and sporadic AD. The limitations of AD mouse models will be discussed and current attempts to overcome these disadvantages will be described. With increasing understanding of the non-neuronal contributions to disease, the development of new methods and models will provide further insights and address important questions regarding the roles of astrocytes and other non-neuronal cells in AD pathophysiology. The next decade will prove to be full of exciting opportunities to address this devastating disease.
Collapse
Affiliation(s)
- Fokion Spanos
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA;
| | - Shane A. Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA;
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
279
|
Zeman T, Balcar VJ, Cahová K, Janoutová J, Janout V, Lochman J, Šerý O. Polymorphism rs11867353 of Tyrosine Kinase Non-Receptor 1 (TNK1) Gene Is a Novel Genetic Marker for Alzheimer's Disease. Mol Neurobiol 2020; 58:996-1005. [PMID: 33070267 DOI: 10.1007/s12035-020-02153-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 09/28/2020] [Indexed: 10/23/2022]
Abstract
Several single-nucleotide polymorphisms (SNPs) and rare variants of non-receptor tyrosine kinase 1 gene (TNK1) have been associated with Alzheimer's disease (AD). To date, none of the associations have proven to be of practical importance in predicting the risk of AD either because the evidence is not conclusive, or the risk alleles occur at very low frequency. In the present study, we are evaluating the associations between rs11867353 polymorphism of TNK1 gene and both AD and mild cognitive impairment (MCI) in a group of 1656 persons. While the association with AD was found to be highly statistically significant (p < 0.0001 for the risk genotype CC), no statistically significant association with MCI could be established. Possible explanation of the apparent discrepancy could be rapid progression of MCI to AD in persons with the CC genotype. Additional findings of the study are statistically significant associations of rs11867353 polymorphism with body mass index, body weight, and body height. The patients with AD and CC genotype had significantly lower values of body mass index and body weight compared with patients with other genotypes. The main outcome of the study is the finding of a previously never described association between the rs11867353 polymorphism of the TNK1 gene and AD. The rs11867353 polymorphism has a potential to become a significant genetic marker when predicting the risk of AD.
Collapse
Affiliation(s)
- Tomáš Zeman
- Laboratory of Neurobiology and Pathological Physiology, Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Veveří 97, 602 00, Brno, Czech Republic
| | - Vladimir J Balcar
- Bosch Institute and Discipline of Anatomy and Histology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Kamila Cahová
- Laboratory of Neurobiology and Molecular Psychiatry, Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Jana Janoutová
- Department of Epidemiology and Public Health, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Vladimír Janout
- Department of Epidemiology and Public Health, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Jan Lochman
- Laboratory of Neurobiology and Pathological Physiology, Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Veveří 97, 602 00, Brno, Czech Republic.,Laboratory of Neurobiology and Molecular Psychiatry, Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Omar Šerý
- Laboratory of Neurobiology and Pathological Physiology, Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, Veveří 97, 602 00, Brno, Czech Republic. .,Laboratory of Neurobiology and Molecular Psychiatry, Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic.
| |
Collapse
|
280
|
Yang Y, Hu S, Lin H, He J, Tang C. Electroacupuncture at GV24 and bilateral GB13 improves cognitive ability via influences the levels of Aβ, p-tau (s396) and p-tau (s404) in the hippocampus of Alzheimer's disease model rats. Neuroreport 2020; 31:1072-1083. [PMID: 32881772 PMCID: PMC7515480 DOI: 10.1097/wnr.0000000000001518] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/26/2020] [Indexed: 12/14/2022]
Abstract
Acupuncture is widely used to treat various neurodegenerative diseases and can effectively improve cognitive and memory states in Alzheimer's disease. However, its mechanism is unclear. We speculated that the effect of acupuncture on cognitive function may be associated with reductions in the levels of Aβ and phosphorylated tau in the brain. In this experiment, 60 male Sprague-Dawley rats were randomly divided into control, model, electroacupuncture and nonacupoint groups. We perform electroacupuncture at Shenting (GV24) and bilateral Benshen (GB13) acupoints once a day for 4 weeks in electroacupuncture group (with 1 day of rest after every 6 days of treatment). The electroacupuncture group showed a better performance in cognitive-related behavior tests and significantly lowers the levels of Aβ, p-tau (s396) and p-tau (s404) in the hippocampus. These results may suggest that electroacupuncture at the GV24 and bilateral GB13 acupoints might improve cognitive functions in Alzheimer's disease by decreasing the levels of Aβ, p-tau (s396) and p-tau (s404) in the brain as these proteins are the main causes of neurological damage and cognitive dysfunction during the pathogenesis underlying Alzheimer's disease.
Collapse
Affiliation(s)
- Yang Yang
- Department of Rehabilitation, Affiliated Jiangmen Traditional Chinese Medicine Hospital of Ji’nan University, Ji’nan University, Jiangmen
| | - Shaowen Hu
- Department of Rehabilitation, Shenzhen Bao’an Traditional Chinese Medicine Hospital Group, Guangzhou University of Chinese Medicine, Shenzhen
| | - Haibo Lin
- Department of Rehabilitation, Affiliated Jiangmen Traditional Chinese Medicine Hospital of Ji’nan University, Ji’nan University, Jiangmen
| | - Jiang He
- College of Acupuncture and Moxibustion, Guangxi University of Chinese Medicine, Guangxi
| | - Chunzhi Tang
- Clinical Medical College of Acupuncture and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
281
|
Adeowo FY, Ejalonibu MA, Elrashedy AA, Lawal MM, Kumalo HM. Multi-target approach for Alzheimer's disease treatment: computational biomolecular modeling of cholinesterase enzymes with a novel 4- N-phenylaminoquinoline derivative reveal promising potentials. J Biomol Struct Dyn 2020; 39:3825-3841. [PMID: 33030113 DOI: 10.1080/07391102.2020.1826129] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The identification of dual inhibitors targeting the active sites of the cholinesterase enzymes, acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE), have lately surfaced as a multi-approach towards Alzheimer treatment. More recently, a novel series of 4-N-phenylaminoquinolines was synthesized and evaluated against AChE and BuChE in which one of the compounds displayed appreciable inhibition compared to the standard compound, galantamine. To provide a clearer picture of the inhibition mechanism of this potent compound at the molecular level, computational biomolecular modeling was carried out. The investigation was initiated with the exploration of the chemical properties of the identified compound 11 b and reference drug, galantamine. Density functional theory (DFT) calculations reveal some conceptual parameters that provide information on the stability and reactivity of the compounds as potential inhibitors. To unveil the binding mechanism, energetics and enzyme-ligand interactions, molecular dynamics (MD) simulations of six different systems were executed over a period. Calculated binding free energy values are in the same order with experimental IC50 data. Identification of the main residues driving optimum binding of the active compound 11 b to the binding region of both AChE and BuChE showed Trp81 and Trp110 as the most important, respectively. It was proposed that the studied compound could serve as a dual inhibitor for AChE and BuChE, therefore, would potentially be a promising moiety in a multi-target approach for the treatment of Alzheimer's disorder.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Fatima Y Adeowo
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa
| | - Murtala A Ejalonibu
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa
| | - Ahmed A Elrashedy
- Molecular Bio-computational and Drug Design Research Group, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Monsurat M Lawal
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa
| | - Hezekiel M Kumalo
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
282
|
He Q, Liu H, Deng S, Chen X, Li D, Jiang X, Zeng W, Lu W. The Golgi Apparatus May Be a Potential Therapeutic Target for Apoptosis-Related Neurological Diseases. Front Cell Dev Biol 2020; 8:830. [PMID: 33015040 PMCID: PMC7493689 DOI: 10.3389/fcell.2020.00830] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/04/2020] [Indexed: 01/04/2023] Open
Abstract
Increasing evidence shows that, in addition to the classical function of protein processing and transport, the Golgi apparatus (GA) is also involved in apoptosis, one of the most common forms of cell death. The structure and the function of the GA is damaged during apoptosis. However, the specific effect of the GA on the apoptosis process is unclear; it may be involved in initiating or promoting apoptosis, or it may inhibit apoptosis. Golgi-related apoptosis is associated with a variety of neurological diseases including glioma, Alzheimer’s disease (AD), Parkinson’s disease (PD), and ischemic stroke. This review summarizes the changes and the possible mechanisms of Golgi structure and function during apoptosis. In addition, we also explore the possible mechanisms by which the GA regulates apoptosis and summarize the potential relationship between the Golgi and certain neurological diseases from the perspective of apoptosis. Elucidation of the interaction between the GA and apoptosis broadens our understanding of the pathological mechanisms of neurological diseases and provides new research directions for the treatment of these diseases. Therefore, we propose that the GA may be a potential therapeutic target for apoptosis-related neurological diseases.
Collapse
Affiliation(s)
- Qiang He
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hui Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Shuwen Deng
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiqian Chen
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Dong Li
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Xuan Jiang
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Wenbo Zeng
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Wei Lu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
283
|
Applications of Hybrid PET/Magnetic Resonance Imaging in Central Nervous System Disorders. PET Clin 2020; 15:497-508. [DOI: 10.1016/j.cpet.2020.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
284
|
Li W, Kui L, Demetrios T, Gong X, Tang M. A Glimmer of Hope: Maintain Mitochondrial Homeostasis to Mitigate Alzheimer's Disease. Aging Dis 2020; 11:1260-1275. [PMID: 33014536 PMCID: PMC7505280 DOI: 10.14336/ad.2020.0105] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 01/05/2020] [Indexed: 12/13/2022] Open
Abstract
Mitochondria are classically known to be cellular energy producers. Given the high-energy demanding nature of neurons in the brain, it is essential that the mitochondrial pool remains healthy and provides a continuous and efficient supply of energy. However, mitochondrial dysfunction is inevitable in aging and neurodegenerative diseases. In Alzheimer’s disease (AD), neurons experience unbalanced homeostasis like damaged mitochondrial biogenesis and defective mitophagy, with the latter promoting the disease-defining amyloid β (Aβ) and p-Tau pathologies impaired mitophagy contributes to inflammation and the aggregation of Aβ and p-Tau-containing neurotoxic proteins. Interventions that restore defective mitophagy may, therefore, alleviate AD symptoms, pointing out the possibility of a novel therapy. This review aims to illustrate mitochondrial biology with a focus on mitophagy and propose strategies to treat AD while maintaining mitochondrial homeostasis.
Collapse
Affiliation(s)
- Wenbo Li
- 1State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, China
| | - Ling Kui
- 2Dana-Farber Cancer Institute, Harvard Medical School, United States
| | | | - Xun Gong
- 4Department of Rheumatology & Immunology, The First Affiliated Hospital of Anhui Medical University, China
| | - Min Tang
- 5Institute of Life Sciences, Jiangsu University, China.,6Center for Innovation in Brain Science, University of Arizona, United States
| |
Collapse
|
285
|
Couly S, Denus M, Bouchet M, Rubinstenn G, Maurice T. Anti-Amnesic and Neuroprotective Effects of Fluoroethylnormemantine in a Pharmacological Mouse Model of Alzheimer's Disease. Int J Neuropsychopharmacol 2020; 24:142-157. [PMID: 32977336 PMCID: PMC7883897 DOI: 10.1093/ijnp/pyaa075] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/15/2020] [Accepted: 09/24/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Current therapies in Alzheimer's disease (AD), including Memantine, have proven to be only symptomatic but not curative or disease modifying. Fluoroethylnormemantine (FENM) is a structural analogue of Memantine, functionalized with a fluorine group that allowed its use as a positron emission tomography tracer. We here analyzed FENM neuroprotective potential in a pharmacological model of AD compared with Memantine. METHODS Swiss mice were treated intracerebroventricularly with aggregated Aβ 25-35 peptide and examined after 1 week in a battery of memory tests (spontaneous alternation, passive avoidance, object recognition, place learning in the water-maze, topographic memory in the Hamlet). Toxicity induced in the mouse hippocampus or cortex was analyzed biochemically or morphologically. RESULTS Both Memantine and FENM showed symptomatic anti-amnesic effects in Aβ 25-35-treated mice. Interestingly, FENM was not amnesic when tested alone at 10 mg/kg, contrarily to Memantine. Drugs injected once per day prevented Aβ 25-35-induced memory deficits, oxidative stress (lipid peroxidation, cytochrome c release), inflammation (interleukin-6, tumor necrosis factor-α increases; glial fibrillary acidic protein and Iba1 immunoreactivity in the hippocampus and cortex), and apoptosis and cell loss (Bcl-2-associated X/B-cell lymphoma 2 ratio; cell loss in the hippocampus CA1 area). However, FENM effects were more robust than observed with Memantine, with significant attenuations vs the Aβ 25-35-treated group. CONCLUSIONS FENM therefore appeared as a potent neuroprotective drug in an AD model, with a superior efficacy compared with Memantine and an absence of direct amnesic effect at higher doses. These results open the possibility to use the compound at more relevant dosages than those actually proposed in Memantine treatment for AD.
Collapse
Affiliation(s)
- Simon Couly
- MMDN, Univ Montpellier, INSERM, EPHE, Montpellier, France
| | - Morgane Denus
- MMDN, Univ Montpellier, INSERM, EPHE, Montpellier, France
| | | | | | - Tangui Maurice
- MMDN, Univ Montpellier, INSERM, EPHE, Montpellier, France,Correspondence: Dr T. Maurice, PhD, INSERM UMR_S1198, Université de Montpellier, cc 105, Place Eugène Bataillon, 34095 Montpellier cedex 5, France ()
| |
Collapse
|
286
|
Jęśko H, Cieślik M, Gromadzka G, Adamczyk A. Dysfunctional proteins in neuropsychiatric disorders: From neurodegeneration to autism spectrum disorders. Neurochem Int 2020; 141:104853. [PMID: 32980494 DOI: 10.1016/j.neuint.2020.104853] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/05/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023]
Abstract
Despite fundamental differences in disease course and outcomes, neurodevelopmental (autism spectrum disorders - ASD) and neurodegenerative disorders (Alzheimer's disease - AD and Parkinson's disease - PD) present surprising, common traits in their molecular pathomechanisms. Uncontrolled oligomerization and aggregation of amyloid β (Aβ), microtubule-associated protein (MAP) tau, or α-synuclein (α-syn) contribute to synaptic impairment and the ensuing neuronal death in both AD and PD. Likewise, the pathogenesis of ASD may be attributed, at least in part, to synaptic dysfunction; attention has also been recently paid to irregularities in the metabolism and function of the Aβ precursor protein (APP), tau, or α-syn. Commonly affected elements include signaling pathways that regulate cellular metabolism and survival such as insulin/insulin-like growth factor (IGF) - PI3 kinase - Akt - mammalian target of rapamycin (mTOR), and a number of key synaptic proteins critically involved in neuronal communication. Understanding how these shared pathomechanism elements operate in different conditions may help identify common targets and therapeutic approaches.
Collapse
Affiliation(s)
- Henryk Jęśko
- Department of Cellular Signalling, M. Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106, Warsaw, Poland.
| | - Magdalena Cieślik
- Department of Cellular Signalling, M. Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106, Warsaw, Poland.
| | - Grażyna Gromadzka
- Cardinal Stefan Wyszynski University, Faculty of Medicine. Collegium Medicum, Wóycickiego 1/3, 01-938, Warsaw, Poland.
| | - Agata Adamczyk
- Department of Cellular Signalling, M. Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106, Warsaw, Poland.
| |
Collapse
|
287
|
Lee C, Willerth SM, Nygaard HB. The Use of Patient-Derived Induced Pluripotent Stem Cells for Alzheimer’s Disease Modeling. Prog Neurobiol 2020; 192:101804. [DOI: 10.1016/j.pneurobio.2020.101804] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 03/31/2020] [Accepted: 04/09/2020] [Indexed: 01/10/2023]
|
288
|
Network Pharmacology Analysis and Molecular Characterization of the Herbal Medicine Formulation Qi-Fu-Yin for the Inhibition of the Neuroinflammatory Biomarker iNOS in Microglial BV-2 Cells: Implication for the Treatment of Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5780703. [PMID: 32952851 PMCID: PMC7481926 DOI: 10.1155/2020/5780703] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/12/2020] [Indexed: 12/23/2022]
Abstract
Aberrant microglial activation drives neuroinflammation and neurodegeneration in Alzheimer's disease (AD). The present study is aimed at investigating whether the herbal formula Qi-Fu-Yin (QFY) could inhibit the inflammatory activation of cultured BV-2 microglia. A network pharmacology approach was employed to predict the active compounds of QFY, protein targets, and affected pathways. The representative pathways and molecular functions of the targets were analyzed by Gene Ontology (GO) and pathway enrichment. A total of 145 active compounds were selected from seven herbal ingredients of QFY. Targets (e.g., MAPT, APP, ACHE, iNOS, and COX-2) were predicted for the selected active compounds based on the relevance to AD and inflammation. As a validation, fractions were sequentially prepared by aqueous extraction, ethanolic precipitation, and HPLC separation, and assayed for downregulating two key proinflammatory biomarkers iNOS and COX-2 in lipopolysaccharide- (LPS-) challenged BV-2 cells by the Western blotting technique. Moreover, the compounds of QFY in 90% ethanol downregulated iNOS in BV-2 cells but showed no activity against COX-2 induction. Among the herbal ingredients of QFY, Angelicae Sinensis Radix and Ginseng Radix et Rhizoma contributed to the selective inhibition of iNOS induction. Furthermore, chemical analysis identified ginsenosides, especially Rg3, as antineuroinflammatory compounds. The herbal formula QFY may ameliorate neuroinflammation via downregulating iNOS in microglia.
Collapse
|
289
|
Lesman-Segev OH, Edwards L, Rabinovici GD. Chronic Traumatic Encephalopathy: A Comparison with Alzheimer's Disease and Frontotemporal Dementia. Semin Neurol 2020; 40:394-410. [PMID: 32820492 DOI: 10.1055/s-0040-1715134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The clinical diagnosis of chronic traumatic encephalopathy (CTE) is challenging due to heterogeneous clinical presentations and overlap with other neurodegenerative dementias. Depending on the clinical presentation, the differential diagnosis of CTE includes Alzheimer's disease (AD), behavioral variant frontotemporal dementia (bvFTD), Parkinson's disease, amyotrophic lateral sclerosis, primary mood disorders, posttraumatic stress disorder, and psychotic disorders. The aim of this article is to compare the clinical aspects, genetics, fluid biomarkers, imaging, treatment, and pathology of CTE to those of AD and bvFTD. A detailed clinical evaluation, neurocognitive assessment, and structural brain imaging can inform the differential diagnosis, while molecular biomarkers can help exclude underlying AD pathology. Prospective studies that include clinicopathological correlations are needed to establish tools that can more accurately determine the cause of neuropsychiatric decline in patients at risk for CTE.
Collapse
Affiliation(s)
- Orit H Lesman-Segev
- Department of Neurology, University of California San Francisco, San Francisco, California
| | - Lauren Edwards
- Department of Neurology, University of California San Francisco, San Francisco, California
| | - Gil D Rabinovici
- Department of Neurology, University of California San Francisco, San Francisco, California.,Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California.,Weill Neuroscience Institute, University of California San Francisco, San Francisco, California
| |
Collapse
|
290
|
Agrawal I, Jha S. Mitochondrial Dysfunction and Alzheimer's Disease: Role of Microglia. Front Aging Neurosci 2020; 12:252. [PMID: 32973488 PMCID: PMC7468434 DOI: 10.3389/fnagi.2020.00252] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/21/2020] [Indexed: 12/14/2022] Open
Abstract
In 1907, Alois Alzheimer observed, as he quoted, development of "numerous fibers" and "adipose saccules" in the brain of his diseased patient Auguste Deter. The neurodegenerative disease became known as Alzheimer's disease (AD) and is the most common cause of dementia worldwide. AD normally develops with aging and is mostly initiated because of the imbalance between the formation and clearance of amyloid-β (Aβ). Formation of neurofibrillary tangles (NFTs) of hyperphosphorylated tau is another hallmark of AD. Neuroinflammation plays a significant role in the development and pathology of AD. This chapter explores the role of mitochondrial dysfunction in microglia in case of AD. Mitochondrial oxidative stress in microglia has been linked to the development of AD. Elevated generation of reactive oxygen species (ROS) and loss of mitochondrial membrane potential through various mechanisms have been observed in AD. Aβ interacts with microglial receptors, such as triggering receptor expressed in myeloid cells 2 (TREM2), activating downstream pathways causing mitochondrial damage and aggravating inflammation and cytotoxicity. Fibrillar Aβ activates nicotinamide adenine dinucleotide phosphate (NADPH) oxidase in microglia leading to elevated induction of mitochondrial ROS which further causes neurotoxicity. Elevated ROS in microglia causes activation of inflammatory and cell death pathways. Production of ATP, regulation of mitochondrial health, autophagy, and mitophagy in microglia play significant roles in the AD pathology. Understanding microglial physiology and mitochondrial dysfunction will enable better therapeutic interventions.
Collapse
Affiliation(s)
- Ishan Agrawal
- Inflammation, Immunity and Tumour Biology Laboratory, Department of Bioscience and Bioengineering, Indian Institute of Technology (IIT) Jodhpur, Jodhpur, India
| | - Sushmita Jha
- Inflammation, Immunity and Tumour Biology Laboratory, Department of Bioscience and Bioengineering, Indian Institute of Technology (IIT) Jodhpur, Jodhpur, India
| |
Collapse
|
291
|
Farr SA, Roesler E, Niehoff ML, Roby DA, McKee A, Morley JE. Metformin Improves Learning and Memory in the SAMP8 Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2020; 68:1699-1710. [PMID: 30958364 DOI: 10.3233/jad-181240] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Metformin is used for the treatment of insulin resistant diabetes. Diabetics are at an increased risk of developing dementia. Recent epidemiological studies suggest that metformin treatment prevents cognitive decline in diabetics. A pilot clinical study found cognitive improvement with metformin in patients with mild cognitive impairment (MCI). Preclinical studies suggest metformin reduces Alzheimer-like pathology in mouse models of Alzheimer's disease (AD). In the current study, we used 11-month-old SAMP8 mice. Mice were given daily injections of metformin at 20 mg/kg/sc or 200 mg/kg/sc for eight weeks. After four weeks, mice were tested in T-maze footshock avoidance, object recognition, and Barnes maze. At the end of the study, brain tissue was collected for analysis of PKC (PKCζ, PKCι, PKCα, PKCγ, PKCɛ), GSK-3β, pGSK-3βser9, pGSK-3βtyr216, pTau404, and APP. Metformin improved both acquisition and retention in SAMP8 mice in T-maze footshock avoidance, retention in novel object recognition, and acquisition in the Barnes maze. Biochemical analysis indicated that metformin increased both atypical and conventional forms of PKC; PKCζ, and PKCα at 20 mg/kg. Metformin significantly increased pGSK-3βser9 at 200 mg/kg, and decreased Aβ at 20 mg/kg and pTau404 and APPc99 at both 20 mg/kg and 200 mg/kg. There were no differences in blood glucose levels between the aged vehicle and metformin treated mice. Metformin improved learning and memory in the SAMP8 mouse model of spontaneous onset AD. Biochemical analysis indicates that metformin improved memory by decreasing APPc99 and pTau. The current study lends support to the therapeutic potential of metformin for AD.
Collapse
Affiliation(s)
- Susan A Farr
- Research and Development Service, VA Medical Center, MO, USA.,Division of Geriatric Medicine, Saint Louis University School of Medicine, MO, USA
| | - Elizabeth Roesler
- Division of Geriatric Medicine, Saint Louis University School of Medicine, MO, USA
| | - Michael L Niehoff
- Division of Geriatric Medicine, Saint Louis University School of Medicine, MO, USA
| | - Deborah A Roby
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, MO, USA
| | - Alexis McKee
- Division of Endocrinology, Diabetes, and Metabolism, Saint Louis University, MO, USA
| | - John E Morley
- Division of Geriatric Medicine, Saint Louis University School of Medicine, MO, USA
| |
Collapse
|
292
|
Frost CV, Zacharias M. From monomer to fibril: Abeta-amyloid binding to Aducanumab antibody studied by molecular dynamics simulation. Proteins 2020; 88:1592-1606. [PMID: 32666627 DOI: 10.1002/prot.25978] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/04/2020] [Accepted: 05/13/2020] [Indexed: 12/27/2022]
Abstract
Alzheimer's disease is one of the most common causes of dementia. It is believed that the aggregation of short Aβ-peptides to form oligomeric and protofibrillar amyloid assemblies plays a central role for disease-relevant neurotoxicity. In recent years, passive immunotherapy has been introduced as a potential treatment strategy with anti-amyloid antibodies binding to Aβ-amyloids and inducing their subsequent degradation by the immune system. Although so far mostly unsuccessful in clinical studies, the high-dosed application of the monoclonal antibody Aducanumab has shown therapeutic potential that might be attributed to its much greater affinity to Aβ-aggregates vs monomeric Aβ-peptides. In order to better understand how Aducanumab interacts with aggregated Aβ-forms compared to monomers, we have generated structural model complexes based on the known structure of Aducanumab in complex with an Aβ2 - 7 -eptitope. Structural models of Aducanumab bound to full-sequence Aβ1 - 40 -monomers, oligomers, protofilaments and mature fibrils were generated and investigated using extensive molecular dynamics simulations to characterize the flexibility and possible additional interactions. Indeed, an aggregate-specific N-terminal binding motif was found in case of Aducanumab binding to oligomers, protofilaments and fibrils that is located next to but not overlapping with the epitope binding site found in the crystal structure with Aβ2 - 7 . Analysis of binding energetics indicates that this motif binds weaker than the epitope but likely contributes to Aducanumab's preference for aggregated Aβ-species. The predicted aggregate-specific binding motif could potentially serve as a basis to reengineer Aducanumab for further enhanced preference to bind Aβ-aggregates vs monomers.
Collapse
Affiliation(s)
- Christina V Frost
- Physics Department T38, Technical University of Munich, Garching, Germany
| | - Martin Zacharias
- Physics Department T38, Technical University of Munich, Garching, Germany
| |
Collapse
|
293
|
Pais M, Martinez L, Ribeiro O, Loureiro J, Fernandez R, Valiengo L, Canineu P, Stella F, Talib L, Radanovic M, Forlenza OV. Early diagnosis and treatment of Alzheimer's disease: new definitions and challenges. REVISTA BRASILEIRA DE PSIQUIATRIA (SAO PAULO, BRAZIL : 1999) 2020; 42:431-441. [PMID: 31994640 PMCID: PMC7430379 DOI: 10.1590/1516-4446-2019-0735] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 11/01/2019] [Indexed: 12/14/2022]
Abstract
The prevalence of Alzheimer's disease (AD), a progressive neurodegenerative disorder, is expected to more than double by 2050. Studies on the pathophysiology of AD have been changing our understanding of this disorder and setting a new scenario for drug development and other therapies. Concepts like the "amyloid cascade" and the "continuum of AD," discussed in this article, are now well established. From updated classifications and recommendations to advances in biomarkers of AD, we aim to critically assess the literature on AD, addressing new definitions and challenges that emerged from recent studies on the subject. Updates on the status of major clinical trials are also given, and future perspectives are discussed.
Collapse
Affiliation(s)
- Marcos Pais
- Laboratório de Neurociências (LIM27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Luana Martinez
- Laboratório de Neurociências (LIM27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Octávio Ribeiro
- Laboratório de Neurociências (LIM27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Júlia Loureiro
- Laboratório de Neurociências (LIM27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Romel Fernandez
- Laboratório de Neurociências (LIM27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Leandro Valiengo
- Laboratório de Neurociências (LIM27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Paulo Canineu
- Laboratório de Neurociências (LIM27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
- Programa de Gerontologia, Pontifícia Universidade Católica de São Paulo (PUC-SP), São Paulo, SP, Brazil
| | - Florindo Stella
- Laboratório de Neurociências (LIM27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
- Instituto de Biociências, Universidade Estadual Paulista (UNESP), Rio Claro, SP, Brazil
| | - Leda Talib
- Laboratório de Neurociências (LIM27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Marcia Radanovic
- Laboratório de Neurociências (LIM27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Orestes V. Forlenza
- Laboratório de Neurociências (LIM27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| |
Collapse
|
294
|
Hoang CL, Ha GH, Pham KTH, Tran BX, Latkin CA, Ho CSH, Ho RCM. Global Mapping of Interventions to Improve Quality of Life of Patients with Alzheimer's Disease during 1990-2018. Dement Geriatr Cogn Disord 2020; 48:221-233. [PMID: 32114583 DOI: 10.1159/000505741] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 01/03/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) with its high burden on disability is known as one of the critical public health issues worldwide. Hence, providing comprehensive care and effective symptomatic treatment are becoming a challenge for many healthcare systems. Quality of life (QOL) has been identified as an important indicator to develop holistic care for people living with AD since it reflects treatment response, the progression of AD, and activities of daily living. This study aimed to identify research trends and landscapes as well as important factors in QOL studies in the field of AD. SUMMARY English research was extracted from the online database Web of Science to analyze research publications' growth rate and content on AD and QOL. VOSviewer was used to visualize the correlations between terms in titles and abstracts. Research topics were created using Latent Dirichlet Allocation of abstracts' content and disciplines. Major landscapes in the QOL assessment included care and treatment for AD in epidemiological studies and clinical trials. Besides, most studies were conducted in high-income countries, such as the USA or the United Kingdom. Findings of our study also identified a lack of contextualized factors and research gaps in terms of QOL among individuals with AD. Key Messages: Further studies should be conducted taking an effort to assess QOL among demented patients as well as advancing knowledge, attitude, and practice among family caregivers.
Collapse
Affiliation(s)
- Chi Linh Hoang
- Institute for Preventive Medicine and Public Health, Hanoi Medical University, Hanoi, Vietnam.,Center of Excellence in Behavioral Medicine, Nguyen Tat Thanh University, Ho Chi Minh City, Vietnam
| | - Giang Hai Ha
- Institute for Global Health Innovations, Duy Tan University, Da Nang, Vietnam, .,Faculty of Pharmacy, Duy Tan University, Da Nang, Vietnam,
| | - Kiet Tuan Huy Pham
- Institute for Preventive Medicine and Public Health, Hanoi Medical University, Hanoi, Vietnam
| | - Bach Xuan Tran
- Institute for Preventive Medicine and Public Health, Hanoi Medical University, Hanoi, Vietnam.,Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Carl A Latkin
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Cyrus S H Ho
- Department of Psychological Medicine, National University Hospital, Singapore, Singapore
| | - Roger C M Ho
- Center of Excellence in Behavioral Medicine, Nguyen Tat Thanh University, Ho Chi Minh City, Vietnam.,Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, Singapore
| |
Collapse
|
295
|
Icaritin Improves Memory and Learning Ability by Decreasing BACE-1 Expression and the Bax/Bcl-2 Ratio in Senescence-Accelerated Mouse Prone 8 (SAMP8) Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:8963845. [PMID: 32714426 PMCID: PMC7345953 DOI: 10.1155/2020/8963845] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 06/03/2020] [Indexed: 12/15/2022]
Abstract
Icaritin (ICT) is the main component in the traditional Chinese herb Epimedium, and it has been shown to have anti-Alzheimer's disease (AD) effects, but its neuroprotective effects and the pharmacological mechanisms are unclear. In the present study, senescence-accelerated mouse prone 8 (SAMP8) mice were randomly divided into a model group and an ICT-treated group. Learning and memory abilities were detected by the Morris water maze assay, and the expression of amyloid beta protein (Aβ) and β-site APP cleavage enzyme 1 (BACE1) was determined by Western blotting and polymerase chain reaction (PCR). Histological changes in CA1 and CA3 were detected by hematoxylin-eosin staining (H&E staining), and the immunohistochemical analysis was used to detect the expression and localization of Bax and Bcl-2. The results showed that compared with the SAMP8 mice, the ICT-treated SAMP8 mice showed improvements in spatial learning and memory retention. In addition, the number of necrotic cells and the morphological changes in CA1 and CA3 areas were significantly alleviated in the group of ICT-treated SAMP8 mice, and the expression of BACE1, Aβ1-42 levels, and the Bax/Bcl-2 ratio in the hippocampus was obviously decreased in the ICT-treated group compared with the control group. The results demonstrated that ICT reduced BACE-1 levels, the contents of Aβ1-42, and the Bax/Bcl-2 ratio, suggesting that ICT might have potential therapeutic benefits by delaying or modifying the progression of AD.
Collapse
|
296
|
Amburana cearensis: Pharmacological and Neuroprotective Effects of Its Compounds. Molecules 2020; 25:molecules25153394. [PMID: 32726999 PMCID: PMC7435960 DOI: 10.3390/molecules25153394] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 07/16/2020] [Accepted: 07/23/2020] [Indexed: 12/11/2022] Open
Abstract
Amburana cearensis A.C. Smith is an endemic tree from Northeastern Brazil used in folk medicine as teas, decocts and syrups for the treatment of various respiratory and inflammatory diseases, since therapeutic properties have been attributed to compounds from its stem bark and seeds. Numerous pharmacological properties of semi-purified extracts and isolated compounds from A. cearensis have been described in several biological systems, ranging from antimicrobial to anti-inflammatory effects. Some of these activities are attributed to coumarins and phenolic compounds, the major compounds present in A. cearensis seed extracts. Multiple lines of research demonstrate these compounds reduce oxidative stress, inflammation and neuronal death induced by glutamate excitotoxicity, events central to most neuropathologies, including Alzheimer’s disease (AD) and Parkinson’s Disease (PD). This review focuses on the botanical aspects, folk medicine use, biological effects and pharmacological activities of A. cearensis compounds and their potential as novel non-toxic drugs for the treatment of neurodegenerative diseases.
Collapse
|
297
|
Allosterism of Nicotinic Acetylcholine Receptors: Therapeutic Potential for Neuroinflammation Underlying Brain Trauma and Degenerative Disorders. Int J Mol Sci 2020; 21:ijms21144918. [PMID: 32664647 PMCID: PMC7404387 DOI: 10.3390/ijms21144918] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 12/21/2022] Open
Abstract
Inflammation is a key physiological phenomenon that can be pervasive when dysregulated. Persistent chronic inflammation precedes several pathophysiological conditions forming one of the critical cellular homeostatic checkpoints. With a steady global surge in inflammatory diseases, it is imperative to delineate underlying mechanisms and design suitable drug molecules targeting the cellular partners that mediate and regulate inflammation. Nicotinic acetylcholine receptors have a confirmed role in influencing inflammatory pathways and have been a subject of scientific scrutiny underlying drug development in recent years. Drugs designed to target allosteric sites on the nicotinic acetylcholine receptors present a unique opportunity to unravel the role of the cholinergic system in regulating and restoring inflammatory homeostasis. Such a therapeutic approach holds promise in treating several inflammatory conditions and diseases with inflammation as an underlying pathology. Here, we briefly describe the potential of cholinergic allosterism and some allosteric modulators as a promising therapeutic option for the treatment of neuroinflammation.
Collapse
|
298
|
Codonopsis pilosula polysaccharide attenuates Aβ toxicity and cognitive defects in APP/PS1 mice. Aging (Albany NY) 2020; 12:13422-13436. [PMID: 32652518 PMCID: PMC7377903 DOI: 10.18632/aging.103445] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 04/20/2020] [Indexed: 12/17/2022]
Abstract
Codonopsis pilosula Polysaccharides (CPPs), a traditional Chinese medicine used for thousands of years, is a potential neuroprotective polysaccharide via a relatively poorly understood mechanism. We previously reported that CPPs attenuated tau pathology in hTau transfected mice and therefore in the current work investigated the effect of CPPs on Aβ toxicity and cognitive defects in APP/PS1 mice model. It was found that one-month intragastric administration of CPPs significantly ameliorated cognitive defects in APP/PS1 mice. In addition, CPPs treatment mitigated the loss of the synaptic plasticity and increased the synaptic proteins including synaptotagmin and PSD95. The expression of Aβ42 and Aβ40 was remarkably decreased in the hippocampus of APP/PS1 mice after CPPs treatment. We also found that CPPs coincubation significantly reduced the amount of APPβ and Aβ42 expression in cells. Intriguingly, the activity of BACE1 was decreased following CPPs treatment in both the hippocampus of APP/PS1 mice and in vitro experiments. Collectively, these results indicated that CPPs attenuated Aβ pathology in APP/PS1 mice, and down-regulating BACE1 might be the underlaying mechanism which could be a therapeutic target for alleviating cognitive defects in AD pathology.
Collapse
|
299
|
Zhang B, Zhao J, Wang Z, Xu L, Liu A, Du G. DL0410 attenuates oxidative stress and neuroinflammation via BDNF/TrkB/ERK/CREB and Nrf2/HO-1 activation. Int Immunopharmacol 2020; 86:106729. [PMID: 32645628 DOI: 10.1016/j.intimp.2020.106729] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/02/2020] [Accepted: 06/19/2020] [Indexed: 12/11/2022]
Abstract
Oxidative stress and neuroinflammation have been deeply associated with Alzheimer's disease. DL0410 is a novel acetylcholinesterase inhibitor with potential anti-oxidative effects in AD-related animal models, while the specific mechanism has not been fully clarified. In this study, DL0410 was predicted to be related to the modification of cell apoptosis, oxidation-reduction process, inflammatory response and ERK1/ERK2 cascade by in silico target fishing and GO enrichment analysis. Then the possible protective effects of DL0410 were evaluated by hydrogen peroxide (H2O2)-induced oxidative stress model and lipopolysaccharides (LPS)-induced neuroinflammation model H2O2 decreased the viability of SH-SY5Y cells, induced malondialdehyde (MDA) accumulation, mitochondrial membrane potential (Δψm) loss and cell apoptosis, which could be reversed by DL0410 dose-dependently, indicating that DL0410 protected SH-SY5Y cells against H2O2-mediated oxidative stress. Western blot analysis showed that DL0410 increased the H2O2-triggered down-regulated TrkB, ERK and CREB phosphorylation and the expression of BDNF. In addition, TrkB inhibitor ANA-12, ERK inhibitor SCH772984 and CREB inhibitor 666-15 eliminated the inhibition of DL0410 on MDA accumulation and Δψm loss. Furthermore, DL0410 attenuates inflammatory responses and ROS production in LPS-treated BV2 cells, which is responsible for Nrf2 and HO-1 up-regulation. The present study demonstrates that DL0410 is a potential activator of the BDNF/TrkB/ERK/CREB and Nrf2/HO-1 pathway and may be a potential candidate for regulating oxidative stress and neuroinflammatory response in the brain. Together, the results showed that DL0410 is a promising drug candidate for treating AD and possibly other nervous system diseases associated with oxidative stress and neuroinflammation.
Collapse
Affiliation(s)
- Baoyue Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jun Zhao
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhe Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lvjie Xu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ailin Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Guanhua Du
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
300
|
Wu Z, Wu H, Sun S, Wu H, Shi W, Song J, Liu J, Zhang Y, Bian F, Jia P, Hou Y. Progesterone attenuates Aβ25–35-induced neuronal toxicity by activating the Ras signalling pathway through progesterone receptor membrane component 1. Life Sci 2020; 253:117360. [DOI: 10.1016/j.lfs.2020.117360] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/16/2020] [Accepted: 01/24/2020] [Indexed: 12/23/2022]
|