201
|
Abstract
Insomnia is an important but widely ignored health problem in modern society. Despite unequivocal evidence on its large prevalence, health and social impacts, comorbidities, and various pharmacologic and nonpharmacologic (behavioral and device-based) approaches, its effective management is still difficult and often incomplete. This article discusses the role of insomnia in modern societies, newer complicating factors, and its overall social and public health burden. Acute insomnia and sleep difficulties during pandemic and confinement are reviewed. The article also focuses on newer developments accumulating in the field of insomnia and possible future trends.
Collapse
Affiliation(s)
- Samson G Khachatryan
- Department of Neurology and Neurosurgery, National Institute of Health, Ministry of Health, Titogradyan 14, Yerevan 0087, Armenia; Sleep and Movement Disorders Center, Somnus Neurology Clinic, Titogradyan 14, Yerevan 0087, Armenia.
| |
Collapse
|
202
|
Fukui K, Kimura S, Kato Y, Kohno M. Effects of far infrared light on Alzheimer's disease-transgenic mice. PLoS One 2021; 16:e0253320. [PMID: 34138944 PMCID: PMC8211253 DOI: 10.1371/journal.pone.0253320] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 06/02/2021] [Indexed: 11/19/2022] Open
Abstract
Far infrared light has been used in many medical procedures. However, the detailed biological mechanisms of infrared light's effects have not yet been elucidated. Many researchers have pointed out the thermal effects of treatments such as infrared saunas, which are known to increase blood flow. Alzheimer's disease (AD) is associated with gradual decreases in brain blood flow and resulting dementia. In this study, we attempted to clarify the beneficial effects of far infrared light using the 5xFAD mouse, a transgenic model of AD. We exposed 5xFAD mice to far infrared light for 5 months. Among the far infrared-exposed AD mice, body weights were significantly decreased, and the levels of nerve growth factor and brain-derived neurotrophic factor protein were significantly increased in selected brain areas (compared to those in non-irradiated AD mice). However, cognition and motor function (as assessed by Morris water maze and Rota Rod tests, respectively) did not differ significantly between the irradiated and non-irradiated AD mouse groups. These results indicated that exposure to far infrared light may have beneficial biological effects in AD mice. However, the experimental schedule and methods may need to be modified to obtain clearer results.
Collapse
Affiliation(s)
- Koji Fukui
- Department of Bioscience and Engineering, Molecular Cell Biology Laboratory, College of Systems Engineering and Sciences, Shibaura Institute of Technology (SIT), Minato, Japan
- Department of Functional Control Systems, Molecular Cell Biology Laboratory, Graduate School of Engineering and Science, SIT, Minato, Japan
- Department of Life Science and Technology, Tokyo Institute of Technology, Meguro, Japan
| | - Shunsuke Kimura
- Department of Bioscience and Engineering, Molecular Cell Biology Laboratory, College of Systems Engineering and Sciences, Shibaura Institute of Technology (SIT), Minato, Japan
| | - Yugo Kato
- Department of Functional Control Systems, Molecular Cell Biology Laboratory, Graduate School of Engineering and Science, SIT, Minato, Japan
| | - Masahiro Kohno
- Department of Life Science and Technology, Tokyo Institute of Technology, Meguro, Japan
- SIT Research Laboratories, The Brain Science & Life Technology Research Center, SIT, Minato, Japan
| |
Collapse
|
203
|
Pérez-Sisqués L, Sancho-Balsells A, Solana-Balaguer J, Campoy-Campos G, Vives-Isern M, Soler-Palazón F, Anglada-Huguet M, López-Toledano MÁ, Mandelkow EM, Alberch J, Giralt A, Malagelada C. RTP801/REDD1 contributes to neuroinflammation severity and memory impairments in Alzheimer's disease. Cell Death Dis 2021; 12:616. [PMID: 34131105 PMCID: PMC8206344 DOI: 10.1038/s41419-021-03899-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 05/25/2021] [Accepted: 06/01/2021] [Indexed: 02/05/2023]
Abstract
RTP801/REDD1 is a stress-regulated protein whose upregulation is necessary and sufficient to trigger neuronal death. Its downregulation in Parkinson's and Huntington's disease models ameliorates the pathological phenotypes. In the context of Alzheimer's disease (AD), the coding gene for RTP801, DDIT4, is responsive to Aβ and modulates its cytotoxicity in vitro. Also, RTP801 mRNA levels are increased in AD patients' lymphocytes. However, the involvement of RTP801 in the pathophysiology of AD has not been yet tested. Here, we demonstrate that RTP801 levels are increased in postmortem hippocampal samples from AD patients. Interestingly, RTP801 protein levels correlated with both Braak and Thal stages of the disease and with GFAP expression. RTP801 levels are also upregulated in hippocampal synaptosomal fractions obtained from murine 5xFAD and rTg4510 mice models of the disease. A local RTP801 knockdown in the 5xFAD hippocampal neurons with shRNA-containing AAV particles ameliorates cognitive deficits in 7-month-old animals. Upon RTP801 silencing in the 5xFAD mice, no major changes were detected in hippocampal synaptic markers or spine density. Importantly, we found an unanticipated recovery of several gliosis hallmarks and inflammasome key proteins upon neuronal RTP801 downregulation in the 5xFAD mice. Altogether our results suggest that RTP801 could be a potential future target for theranostic studies since it could be a biomarker of neuroinflammation and neurotoxicity severity of the disease and, at the same time, a promising therapeutic target in the treatment of AD.
Collapse
Affiliation(s)
- Leticia Pérez-Sisqués
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Anna Sancho-Balsells
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Júlia Solana-Balaguer
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Genís Campoy-Campos
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Marcel Vives-Isern
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Ferran Soler-Palazón
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Marta Anglada-Huguet
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- CAESAR Research Center, Bonn, Germany
| | | | - Eva-Maria Mandelkow
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- CAESAR Research Center, Bonn, Germany
| | - Jordi Alberch
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Catalonia, Spain
| | - Albert Giralt
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
- Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Catalonia, Spain.
| | - Cristina Malagelada
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Catalonia, Spain.
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
204
|
Lazar RM, Howard VJ, Kernan WN, Aparicio HJ, Levine DA, Viera AJ, Jordan LC, Nyenhuis DL, Possin KL, Sorond FA, White CL. A Primary Care Agenda for Brain Health: A Scientific Statement From the American Heart Association. Stroke 2021; 52:e295-e308. [PMID: 33719523 PMCID: PMC8995075 DOI: 10.1161/str.0000000000000367] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A healthy brain is critical for living a longer and fuller life. The projected aging of the population, however, raises new challenges in maintaining quality of life. As we age, there is increasing compromise of neuronal activity that affects functions such as cognition, also making the brain vulnerable to disease. Once pathology-induced decline begins, few therapeutic options are available. Prevention is therefore paramount, and primary care can play a critical role. The purpose of this American Heart Association scientific statement is to provide an up-to-date summary for primary care providers in the assessment and modification of risk factors at the individual level that maintain brain health and prevent cognitive impairment. Building on the 2017 American Heart Association/American Stroke Association presidential advisory on defining brain health that included "Life's Simple 7," we describe here modifiable risk factors for cognitive decline, including depression, hypertension, physical inactivity, diabetes, obesity, hyperlipidemia, poor diet, smoking, social isolation, excessive alcohol use, sleep disorders, and hearing loss. These risk factors include behaviors, conditions, and lifestyles that can emerge before adulthood and can be routinely identified and managed by primary care clinicians.
Collapse
|
205
|
Wang L, Jing R, Wang X, Wang B, Guo K, Zhao J, Gao S, Xu N, Xuan X. A method for the expression of fibroblast growth factor 14 and assessment of its neuroprotective effect in an Alzheimer's disease model. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:994. [PMID: 34277794 PMCID: PMC8267273 DOI: 10.21037/atm-21-2492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/11/2021] [Indexed: 11/29/2022]
Abstract
Background Fibroblast growth factor (FGF) 14 is a member of the FGF family that is mainly expressed in the central nervous system. FGF14 has a close association with the occurrence of neurodegenerative conditions; however, its significance in Alzheimer’s disease (AD) has yet to be evaluated. Therefore, we sought to obtain a large amount of exogenous FGF14 protein and explore its effect in a cellular model of AD. Methods FGF14 protein was expressed in an Escherichia coli system using gene recombination technology. Purified protein was obtained through washing and renaturation of inclusion bodies combined with nickel column affinity chromatography. The AD model was established via Aβ25-35-induced injury in PC12 cells. Changes in the levels of lactate dehydrogenase and malondialdehyde were detected, and the neuroprotective effect of recombinant human FGF14 (rhFGF14) was evaluated through double-fluorescence staining and flow cytometry apoptosis detection. For further exploration of rhFGF14-mediated regulation of mitogen-activated protein kinase (MAPK) signaling, western blot was employed. Results We successfully induced large amounts of insoluble rhFGF14. Following solubilization and refolding of the rhFGF14 from inclusion bodies, high purity rhFGF14 was purified by Nickel affinity column chromatography. The results showed that rhFGF14 alleviated Aβ25-3-induced PC12 cell injury by inhibiting the phosphorylation of p38, extracellular signal-regulated kinase 1/2, and c-Jun N-terminal kinase, thus suppressing the MAPK signaling pathway. Conclusions FGF14 performed a neuroprotective role in our in vitro AD model via its inhibition of MAPK signaling, highlighting its potential as a therapeutic drug for neurodegenerative conditions.
Collapse
Affiliation(s)
- Lusheng Wang
- Department of Dermatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Rongrong Jing
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Xing Wang
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Baohui Wang
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Keke Guo
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Jungang Zhao
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Shuang Gao
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Nuo Xu
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Xuan Xuan
- Department of Dermatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
206
|
Weng G, Zhou B, Liu T, Huang Z, Huang S. Tetramethylpyrazine Improves Cognitive Function of Alzheimer's Disease Mice by Regulating SSTR4 Ubiquitination. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:2385-2399. [PMID: 34103899 PMCID: PMC8179737 DOI: 10.2147/dddt.s290030] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 04/12/2021] [Indexed: 01/08/2023]
Abstract
Purpose Many researches have investigated the functions of tetramethylpyrazine (TMP) in Alzheimer's disease (AD). This study aimed to discuss the underlying mechanism of TMP in AD mice. Methods TMP (200 mg/kg) was administered to 6-month-old APP/PS1 transgenic mice, and behavioral changes and hippocampal nerve injury in AD mice were detected. Apoptosis and autophagy-related protein levels were detected. Changes in gene expression before and after TMP treatment were compared using transcriptome sequencing. The effects of Cullin 4B (CUL4B) overexpression and somatostatin receptor 4 (SSTR4) silencing on AD symptoms and SSTR4 ubiquitination in APP/PS1 mice were observed. SH-SY5Y and PC12 cells were treated with 25 μmol/L Aβ25-35 and TMP to observe cell viability, apoptosis, and autophagy. Cell viability and apoptosis were measured again after treatment with proteasome inhibitor MG132 or lysosomal inhibitor 3-mA. Results TMP treatment improved the behavioral cognition of APP/PS1 mice and improved the neuronal apoptosis and damage in brain tissue. CUL4B was significantly upregulated in APP/PS1 mouse brain tissue, and SSRT4 protein was downregulated, and the levels of CUL4B and SSRT4 were negatively correlated. TMP treatment downregulated CUL4B, inhibited SSRT4 ubiquitination and upregulated SSRT4 protein level in APP/PS1 mouse brain tissue, while CUL4B overexpression or SSRT4 silencing reversed the effect of TMP. TMP and MG132 improved the decreased activity, increased apoptosis and increased SSRT4 protein in SH-SY5Y and PC12 cells treated with Aβ25-35, but not 3-mA. CUL4B overexpression promoted the ubiquitination of SSTR4 in cells, which partially reversed the effect of TMP. Conclusion TMP could improve the cognitive ability of AD mice by inhibiting CUL4B expression and the ubiquitination degradation of SSTR, and alleviating neuronal apoptosis and injury. This study may offer a new therapeutic option for AD treatment.
Collapse
Affiliation(s)
- Guohu Weng
- Department of Encephalopathy, Hainan Provincial Hospital of Traditional Chinese Medicine, Haikou, 570203, Hainan, People's Republic of China
| | - Bo Zhou
- Department of Cardiology, Hainan Provincial Hospital of Traditional Chinese Medicine, Haikou, 570203, Hainan, People's Republic of China
| | - Tao Liu
- Department of Neurology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, People's Republic of China
| | - Zhengxin Huang
- Department of Cardiology, Hainan Provincial Hospital of Traditional Chinese Medicine, Haikou, 570203, Hainan, People's Republic of China
| | - Shixiong Huang
- Department of Neurology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, People's Republic of China
| |
Collapse
|
207
|
Strand H, Garabet L, Bjelke B, Sithiravel C, Hardang IM, Moe MK. β-Amyloid in Cerebrospinal Fluid: How to Keep It Floating (Not Sticking) by Standardization of Preanalytic Processes and Collection Tubes. J Appl Lab Med 2021; 6:1155-1164. [PMID: 34059876 DOI: 10.1093/jalm/jfab024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 12/15/2020] [Indexed: 11/13/2022]
Abstract
BACKGROUND Phosphorylated tau (pTau), total tau (tTau), and β-amyloid (Aβ) are established cerebrospinal fluid (CSF) biomarkers used to help diagnose Alzheimer disease. Preanalytic workups of CSF samples lack harmonization, making interlaboratory comparison of these biomarkers challenging. The Aβ adsorbs to sample tubes, yielding underestimated concentrations, and may result in false Alzheimer disease diagnosis. Our primary aim was to compare Aβ recovery across multiple polypropylene tubes and to test the stability of tTau, pTau, and Aβ in the best performing tube. METHODS Eight polypropylene tubes were tested using 3 CSF pools with Aβ concentrations <500, 500-1000, and >1000 ng/L. All samples were analyzed in duplicate. Tubes were cut open to assess their different infrared adsorption spectra. Freshly drawn CSF from 14 patients was distributed into 4 Sarstedt 5-mL (no. 63.504.027; Sar5CSF) tubes, left at room temperature for up to 7 days, and analyzed for pTau, tTau, and Aβ by ELISA. RESULTS Two Sarstedt 5-mL tubes and a Sarstedt 10-mL (Sar10CSF) tube showed significantly higher Aβ recovery at all 3 concentrations compared with the 5 other tubes. The infrared adsorption spectra of Sar10CSF and Sar5CSF tubes were practically identical, unlike the other tubes. No significant loss of pTau, tTau, and Aβ was observed in CSF left at room temperature for up to 7 days (P > 0.05). CONCLUSIONS Recovery of Aβ from Sar5CSF tubes is equivalent to Aβ recovery from Sar10CSF tubes. Levels of pTau, tTau, and Aβ were stable for at least 7 days at room temperature but not at 37 °C.
Collapse
Affiliation(s)
- Heidi Strand
- Multidisciplinary Laboratory Medicine and Medical Biochemistry, Akershus University Hospital, Lørenskog, Norway
| | - Lamya Garabet
- Multidisciplinary Laboratory Medicine and Medical Biochemistry, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Research, Østfold Hospital Trust, Grålum, Norway
| | - Börje Bjelke
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Neurology, Akershus University Hospital, Lørenskog, Norway
| | - Cindhya Sithiravel
- Multidisciplinary Laboratory Medicine and Medical Biochemistry, Akershus University Hospital, Lørenskog, Norway
| | - Ingrid Marie Hardang
- Multidisciplinary Laboratory Medicine and Medical Biochemistry, Akershus University Hospital, Lørenskog, Norway
| | - Morten K Moe
- Multidisciplinary Laboratory Medicine and Medical Biochemistry, Akershus University Hospital, Lørenskog, Norway
| |
Collapse
|
208
|
Kubis-Kubiak A, Wiatrak B, Piwowar A. The Impact of High Glucose or Insulin Exposure on S100B Protein Levels, Oxidative and Nitrosative Stress and DNA Damage in Neuron-Like Cells. Int J Mol Sci 2021; 22:ijms22115526. [PMID: 34073816 PMCID: PMC8197274 DOI: 10.3390/ijms22115526] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 12/25/2022] Open
Abstract
Alzheimer’s disease (AD) is attracting considerable interest due to its increasing number of cases as a consequence of the aging of the global population. The mainstream concept of AD neuropathology based on pathological changes of amyloid β metabolism and the formation of neurofibrillary tangles is under criticism due to the failure of Aβ-targeting drug trials. Recent findings have shown that AD is a highly complex disease involving a broad range of clinical manifestations as well as cellular and biochemical disturbances. The past decade has seen a renewed importance of metabolic disturbances in disease-relevant early pathology with challenging areas in establishing the role of local micro-fluctuations in glucose concentrations and the impact of insulin on neuronal function. The role of the S100 protein family in this interplay remains unclear and is the aim of this research. Intracellularly the S100B protein has a protective effect on neurons against the toxic effects of glutamate and stimulates neurites outgrowth and neuronal survival. At high concentrations, it can induce apoptosis. The aim of our study was to extend current knowledge of the possible impact of hyper-glycemia and -insulinemia directly on neuronal S100B secretion and comparison to oxidative stress markers such as ROS, NO and DBSs levels. In this paper, we have shown that S100B secretion decreases in neurons cultured in a high-glucose or high-insulin medium, while levels in cell lysates are increased with statistical significance. Our findings demonstrate the strong toxic impact of energetic disturbances on neuronal metabolism and the potential neuroprotective role of S100B protein.
Collapse
Affiliation(s)
- Adriana Kubis-Kubiak
- Department of Toxicology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland;
- Correspondence:
| | - Benita Wiatrak
- Department of Pharmacology, Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland;
| | - Agnieszka Piwowar
- Department of Toxicology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland;
| |
Collapse
|
209
|
Virtual Reality-Based Cognitive Stimulation on People with Mild to Moderate Dementia due to Alzheimer's Disease: A Pilot Randomized Controlled Trial. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18105290. [PMID: 34065698 PMCID: PMC8156930 DOI: 10.3390/ijerph18105290] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/09/2021] [Accepted: 05/12/2021] [Indexed: 12/02/2022]
Abstract
The use of ecologically oriented approaches with virtual reality (VR) depicting instrumental activities of daily living (IADL) is a promising approach for interventions on acquired brain injuries. However, the results of such an approach on dementia caused by Alzheimer’s disease (AD) are still lacking. This research reports on a pilot randomized controlled trial that aimed to explore the effect of a cognitive stimulation reproducing several IADL in VR on people with mild-to-moderate dementia caused by AD. Patients were recruited from residential care homes of Santa Casa da Misericórdia da Amadora (SCMA), which is a relevant nonprofit social and healthcare provider in Portugal. This intervention lasted two months, with a total of 10 sessions (two sessions/week). A neuropsychological assessment was carried out at the baseline and follow-up using established neuropsychological instruments for assessing memory, attention, and executive functions. The sample consisted of 17 patients of both genders randomly assigned to the experimental and control groups. The preliminary results suggested an improvement in overall cognitive function in the experimental group, with an effect size corresponding to a large effect in global cognition, which suggests that this approach is effective for neurocognitive stimulation in older adults with dementia, contributing to maintaining cognitive function in AD.
Collapse
|
210
|
Wu G, Geng H, Xu R, Deng M, Yang C, Xun C, Wang Y, Cai Q, Chen P. Preparation of a CaTiO 3/Al 3+/Pr 3+/Sm 3+ nanocomposite for enrichment of exosomes in human serum. Talanta 2021; 226:122186. [PMID: 33676717 DOI: 10.1016/j.talanta.2021.122186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/28/2021] [Accepted: 01/30/2021] [Indexed: 10/22/2022]
Abstract
Exosomes (30-200 nm) play important roles in intercellular communication. Because their contents differ between healthy individuals and subjects diagnosed with various diseases, exosomes have been regarded as potential sources of biomarkers for clinical diagnosis. However, the accuracy of diagnosis by exosomal biomarkers is highly dependent on the extraction efficiency, yield, and the quality of exosomes. Hence, inexpensive, convenient, and fast exosome separation methods are required. In the present study, the CaTiO3/Al3+/Pr3+/Sm3+ nanocomposite was synthesized and applied in highly selective and efficient separation of exosomes. Notably, the developed material exhibited higher specificity and efficiency than commercially available TiO2. Moreover, CaTiO3/Al3+/Pr3+/Sm3+ could be reused at least three times without any significant decrease in efficiency. The synthesized material was also used for the extraction of exosomes from the serums of patients with Alzheimer's disease (AD) and healthy controls. The exosomes were subjected to two-dimensional gel electrophoresis (2-DE) separation and matrix-assisted laser desorption/ionization-time of flight (MALDI TOF/TOF) mass spectrometry analysis. It was found that five proteins in the exosomes were evidently upregulated, while one protein was downregulated. Among the detected proteins, serum amyloid P-component (SAP) has been reported to be closely related to pathogenesis of AD. The obtained results indicated that the developed method involving separation and analysis of serum exosomes could be used for disease diagnosis or postoperative clinical monitoring.
Collapse
Affiliation(s)
- Guangyao Wu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Hongchao Geng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China
| | - Rongfang Xu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Min Deng
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Changcheng Yang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Chengfeng Xun
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Ying Wang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China; State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Qingyun Cai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China.
| | - Ping Chen
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China; State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China.
| |
Collapse
|
211
|
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 DOI: 10.2174/1381612826666201125112131] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [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, United States
| |
Collapse
|
212
|
Design, synthesis and evaluation of cholinesterase hybrid inhibitors using a natural steroidal alkaloid as precursor. Bioorg Chem 2021; 111:104893. [PMID: 33882364 DOI: 10.1016/j.bioorg.2021.104893] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 04/01/2021] [Accepted: 04/03/2021] [Indexed: 01/05/2023]
Abstract
To date, Alzheimer's disease is the most alarming neurodegenerative disorder worldwide. This illness is multifactorial in nature and cholinesterase inhibitors have been the ones used in clinical treatments. In this context, many of these drugs selectively inhibit the acetylcholinesterase enzyme interacting in both the active site and the peripheric anionic site. Besides, some agents have exhibited extensive benefits being able to co-inhibit butyrylcholinesterase. In this contribution, a strategy previously explored by numerous authors is reported; the synthesis of hybrid cholinesterase inhibitors. This strategy uses a molecule of recognized high inhibitory activity (tacrine) together with a steroidal alkaloid of natural origin using different connectors. The biological assays demonstrated the improvement in the inhibitory activity compared to the alkaloidal precursor, together with the reinforcement of the interactions in multiple sites of the enzymatic cavity. This strategy should be explored and exploited in this area. Docking and molecular dynamic studies were performed to explain enzyme-ligand interactions, assisting a structure-activity relationship analysis.
Collapse
|
213
|
Tomino C, Ilari S, Solfrizzi V, Malafoglia V, Zilio G, Russo P, Proietti S, Marcolongo F, Scapagnini G, Muscoli C, Rossini PM. Mild Cognitive Impairment and Mild Dementia: The Role of Ginkgo biloba (EGb 761 ®). Pharmaceuticals (Basel) 2021; 14:ph14040305. [PMID: 33915701 PMCID: PMC8065464 DOI: 10.3390/ph14040305] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/15/2021] [Accepted: 03/26/2021] [Indexed: 12/17/2022] Open
Abstract
Mild cognitive impairment (MCI) and dementia are clinically prevalent in the elderly. There is a high risk of cognitive decline in patients diagnosed with MCI or dementia. This review describes the effectiveness of Ginkgo biloba leaf special extract EGb 761® for the treatment of dementia syndromes and EGb 761® combination therapy with other medications for symptomatic dementia. This drug has shown convincing results, improving cognitive function, neuropsychiatric symptoms and consequent reduction of caregiver stress and maintenance of autonomy in patients with age-related cognitive decline, MCI and mild to moderate dementia. Currently, there is little evidence to support the combination therapy with anti-dementia drugs and, therefore, more evidence is needed to evaluate the role of EGb 761® in mixed therapy.
Collapse
Affiliation(s)
- Carlo Tomino
- Scientific Direction, IRCCS San Raffaele Roma, 00166 Rome, Italy; (C.T.); (S.P.)
| | - Sara Ilari
- Department of Health Science, Institute of Research for Food Safety & Health (IRC-FSH), University “Magna Graecia” of Catanzaro, 88201 Catanzaro, Italy; (S.I.); (C.M.)
| | - Vincenzo Solfrizzi
- Clinica Medica “Frugoni” and Geriatric Medicine-Memory Unit, University of Bari Aldo Moro, 70122 Bari, Italy;
| | - Valentina Malafoglia
- Institute for Research on Pain, ISAL Foundation, Torre Pedrera, 47922 Rimini, Italy;
| | - Guglielmo Zilio
- Scientific Department, Schwabe Pharma Italia S.r.l., 39044 Egna, Italy;
| | - Patrizia Russo
- Clinical and Molecular Epidemiology, IRCCS San Raffaele Roma, 00166 Rome, Italy;
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, Via di Val Cannuta, 247, 00166 Rome, Italy
- Correspondence: or
| | - Stefania Proietti
- Scientific Direction, IRCCS San Raffaele Roma, 00166 Rome, Italy; (C.T.); (S.P.)
| | - Federica Marcolongo
- Clinical and Molecular Epidemiology, IRCCS San Raffaele Roma, 00166 Rome, Italy;
| | - Giovanni Scapagnini
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy;
| | - Carolina Muscoli
- Department of Health Science, Institute of Research for Food Safety & Health (IRC-FSH), University “Magna Graecia” of Catanzaro, 88201 Catanzaro, Italy; (S.I.); (C.M.)
| | - Paolo Maria Rossini
- Department of Neuroscience & Neurorehabilitation, IRCCS San Raffaele Roma, 00163 Rome, Italy;
| |
Collapse
|
214
|
Paulus A, Engdahl A, Yang Y, Boza-Serrano A, Bachiller S, Torres-Garcia L, Svanbergsson A, Garcia MG, Gouras GK, Li JY, Deierborg T, Klementieva O. Amyloid Structural Changes Studied by Infrared Microspectroscopy in Bigenic Cellular Models of Alzheimer's Disease. Int J Mol Sci 2021; 22:3430. [PMID: 33810433 PMCID: PMC8037084 DOI: 10.3390/ijms22073430] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease affects millions of lives worldwide. This terminal disease is characterized by the formation of amyloid aggregates, so-called amyloid oligomers. These oligomers are composed of β-sheet structures, which are believed to be neurotoxic. However, the actual secondary structure that contributes most to neurotoxicity remains unknown. This lack of knowledge is due to the challenging nature of characterizing the secondary structure of amyloids in cells. To overcome this and investigate the molecular changes in proteins directly in cells, we used synchrotron-based infrared microspectroscopy, a label-free and non-destructive technique available for in situ molecular imaging, to detect structural changes in proteins and lipids. Specifically, we evaluated the formation of β-sheet structures in different monogenic and bigenic cellular models of Alzheimer's disease that we generated for this study. We report on the possibility to discern different amyloid signatures directly in cells using infrared microspectroscopy and demonstrate that bigenic (amyloid-β, α-synuclein) and (amyloid-β, Tau) neuron-like cells display changes in β-sheet load. Altogether, our findings support the notion that different molecular mechanisms of amyloid aggregation, as opposed to a common mechanism, are triggered by the specific cellular environment and, therefore, that various mechanisms lead to the development of Alzheimer's disease.
Collapse
Affiliation(s)
- Agnes Paulus
- Medical Microspectroscopy Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (A.P.); (A.E.)
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (Y.Y.); (A.B.-S.); (S.B.); (M.G.G.)
| | - Anders Engdahl
- Medical Microspectroscopy Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (A.P.); (A.E.)
| | - Yiyi Yang
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (Y.Y.); (A.B.-S.); (S.B.); (M.G.G.)
| | - Antonio Boza-Serrano
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (Y.Y.); (A.B.-S.); (S.B.); (M.G.G.)
| | - Sara Bachiller
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (Y.Y.); (A.B.-S.); (S.B.); (M.G.G.)
| | - Laura Torres-Garcia
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (L.T.-G.); (G.K.G.)
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (A.S.); (J.-Y.L.)
| | - Alexander Svanbergsson
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (A.S.); (J.-Y.L.)
| | - Megg G. Garcia
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (Y.Y.); (A.B.-S.); (S.B.); (M.G.G.)
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (L.T.-G.); (G.K.G.)
| | - Gunnar K. Gouras
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (L.T.-G.); (G.K.G.)
| | - Jia-Yi Li
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (A.S.); (J.-Y.L.)
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (Y.Y.); (A.B.-S.); (S.B.); (M.G.G.)
| | - Oxana Klementieva
- Medical Microspectroscopy Laboratory, Department of Experimental Medical Science, Lund University, 22184 Lund, Sweden; (A.P.); (A.E.)
- Lund Institute for Advanced Neutron and X-ray Science (LINXS), 22370 Lund, Sweden
| |
Collapse
|
215
|
Butler CA, Popescu AS, Kitchener EJA, Allendorf DH, Puigdellívol M, Brown GC. Microglial phagocytosis of neurons in neurodegeneration, and its regulation. J Neurochem 2021; 158:621-639. [PMID: 33608912 DOI: 10.1111/jnc.15327] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/13/2021] [Accepted: 02/10/2021] [Indexed: 02/06/2023]
Abstract
There is growing evidence that excessive microglial phagocytosis of neurons and synapses contributes to multiple brain pathologies. RNA-seq and genome-wide association (GWAS) studies have linked multiple phagocytic genes to neurodegenerative diseases, and knock-out of phagocytic genes has been found to protect against neurodegeneration in animal models, suggesting that excessive microglial phagocytosis contributes to neurodegeneration. Here, we review recent evidence that microglial phagocytosis of live neurons and synapses causes neurodegeneration in animal models of Alzheimer's disease and other tauopathies, Parkinson's disease, frontotemporal dementias, multiple sclerosis, retinal degeneration and neurodegeneration induced by ischaemia, infection or ageing. We also review factors regulating microglial phagocytosis of neurons, including: nucleotides, frackalkine, phosphatidylserine, calreticulin, UDP, CD47, sialylation, complement, galectin-3, Apolipoprotein E, phagocytic receptors, Siglec receptors, cytokines, microglial epigenetics and expression profile. Some of these factors may be potential treatment targets to prevent neurodegeneration mediated by excessive microglial phagocytosis of live neurons and synapses.
Collapse
Affiliation(s)
- Claire A Butler
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Alma S Popescu
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | | | - Mar Puigdellívol
- Department of Biochemistry, University of Cambridge, Cambridge, UK.,Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
216
|
Khurana K, Kumar M, Bansal N. Lacidipine Prevents Scopolamine-Induced Memory Impairment by Reducing Brain Oxido-nitrosative Stress in Mice. Neurotox Res 2021; 39:1087-1102. [PMID: 33721210 DOI: 10.1007/s12640-021-00346-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/08/2021] [Accepted: 02/28/2021] [Indexed: 02/06/2023]
Abstract
Cholinergic deficits and oxido-nitrosative stress are consistently associated with Alzheimer's disease (AD). Previous findings indicate that acetylcholine subdues Ca2+ current in the brain. Cholinergic antagonists (e.g., scopolamine) can instigate Ca2+-induced redox imbalance, inflammation, and cell-death pathways leading to AD-type memory impairment. Earlier, several Ca2+-channel blockers (CCB, e.g., dihydropyridine type) or cholinergic enhancers showed promising results in animal models of AD. In the present research, pretreatment effects of lacidipine (L-type CCB) on learning and memory functions were investigated using the scopolamine mouse model of AD. Swiss albino mice (20-25 g) were administered lacidipine (1 and 3 mg/kg) for 14 days. Scopolamine, an anti-muscarinic drug, was given (1 mg/kg) from days 8 to 14. The mice were subjected to elevated plus maze (EPM) and passive-avoidance (PA) paradigms. Bay-K8644 (a Ca2+-channel agonist) was administered before behavioral studies on days 13 and 14. Biochemical parameters of oxidative stress and acetylcholinesterase (AChE) activity were quantified using the whole brain. Behavioral studies showed an increase in transfer latency (TL) in the EPM test and a decrease in step-through latency (STL) in the PA test in scopolamine-administered mice. Scopolamine enhanced the AChE activity and oxidative stress in the brain of mice which resulted in memory impairment. Lacidipine prevented the amnesia against scopolamine and reduced the oxidative stress and AChE activity in the brain of mice. Bay-K8644 attenuated the lacidipine-induced improvement in memory and redox balance in scopolamine-administered mice. Lacidipine can prevent the oxidative stress and improve the cholinergic function in the brain. These properties of lacidipine can mitigate the pathogenesis of AD-type dementia.
Collapse
Affiliation(s)
- Kunal Khurana
- I.K. Gujral Punjab Technical University, Kapurthala, Punjab, 144603, India.,Department of Pharmacology, Amar Shaheed Baba Ajeet Singh Jujhar Singh Memorial College of Pharmacy, Bela, Ropar, Punjab, 140111, India
| | - Manish Kumar
- Department of Pharmacology, Amar Shaheed Baba Ajeet Singh Jujhar Singh Memorial College of Pharmacy, Bela, Ropar, Punjab, 140111, India.,Chitkara College of Pharmacy, Chitkara University, Punjab, 140111, India
| | - Nitin Bansal
- Department of Pharmacology, Amar Shaheed Baba Ajeet Singh Jujhar Singh Memorial College of Pharmacy, Bela, Ropar, Punjab, 140111, India. .,Department of Pharmaceutical Sciences, Chaudhary Bansi Lal University (CBLU), Bhiwani, Haryana, 127021, India.
| |
Collapse
|
217
|
Derby CA, Hutchins F, Greendale GA, Matthews KA, Sternfeld B, Everson-Rose SA, Kazlauskaite R, Whitmer RA, Brooks MM. Cardiovascular risk and midlife cognitive decline in the Study of Women's Health Across the Nation. Alzheimers Dement 2021; 17:1342-1352. [PMID: 33710770 DOI: 10.1002/alz.12300] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/01/2020] [Accepted: 01/02/2021] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Cardiovascular risk factors in midlife have been linked to late life risk for Alzheimer's disease and related dementias (ADRD). The relation of vascular risk factors on cognitive decline within midlife has been less studied. METHODS Using data from the Study of Women's Health Across the Nation, we examined associations of midlife hypertension, elevated lipid levels, diabetes, fasting glucose, central adiposity, and Framingham heart age with rates of cognitive decline in women who completed multiple cognitive assessments of processing speed, and working and verbal memory during midlife. RESULTS Diabetes, elevated fasting glucose, central obesity, and heart age greater than chronological age were associated with rate of decline in processing speed during midlife. Vascular risk factors were not related to rate of decline in working or verbal memory. DISCUSSION Midlife may be a critical period for intervening on cardiovascular risk factors to prevent or delay later life cognitive impairment and ADRD.
Collapse
Affiliation(s)
- Carol A Derby
- Saul R. Korey Department of Neurology, Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Franya Hutchins
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Gail A Greendale
- Division of Geriatrics, University of California at Los Angeles, Los Angeles, California, USA
| | - Karen A Matthews
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Susan A Everson-Rose
- Department of Medicine and Program in Health Disparities Research, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rasa Kazlauskaite
- Division of Endocrinology and Metabolism, Rush University Medical Center, Chicago, Illinois, USA
| | - Rachel A Whitmer
- Division of Epidemiology, Department of Public Health Sciences, School of Medicine, University of California, Davis, Davis, California, USA
| | - Maria M Brooks
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
218
|
Huang SH, Fang ST, Chen YC. Molecular Mechanism of Vitamin K2 Protection against Amyloid-β-Induced Cytotoxicity. Biomolecules 2021; 11:423. [PMID: 33805625 PMCID: PMC8000266 DOI: 10.3390/biom11030423] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/05/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
The pathological role of vitamin K2 in Alzheimer's disease (AD) involves a definite link between impaired cognitive functions and decreased serum vitamin K levels. Vitamin K2 supplementation may have a protective effect on AD. However, the mechanism underlying vitamin K2 protection has not been elucidated. With the amyloid-β (Aβ) cascade hypothesis, we constructed a clone containing the C-terminal fragment of amyloid precursor protein (β-CTF/APP), transfected in astroglioma C6 cells and used this cell model (β-CTF/C6) to study the protective effect of vitamin K2 against Aβ cytotoxicity. Both cellular and biochemical assays, including cell viability and reactive oxygen species (ROS), assays assay, and Western blot and caspase activity analyses, were used to characterize and unveil the protective role and mechanism of vitamin K2 protecting against Aβ-induced cytotoxicity. Vitamin K2 treatment dose-dependently decreased the death of neural cells. The protective effect of vitamin K2 could be abolished by adding warfarin, a vitamin K2 antagonist. The addition of vitamin K2 reduced the ROS formation and inhibited the caspase-3 mediated apoptosis induced by Aβ peptides, indicating that the mechanism underlying the vitamin K2 protection is likely against Aβ-mediated apoptosis. Inhibitor assay and Western blot analyses revealed that the possible mechanism of vitamin K2 protection against Aβ-mediated apoptosis might be via regulating phosphatidylinositol 3-kinase (PI3K) associated-signaling pathway and inhibiting caspase-3-mediated apoptosis. Our study demonstrates that vitamin K2 can protect neural cells against Aβ toxicity.
Collapse
Affiliation(s)
| | | | - Yi-Cheng Chen
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan; (S.-H.H.); (S.-T.F.)
| |
Collapse
|
219
|
Wang W, Zhou Q, Jiang T, Li S, Ye J, Zheng J, Wang X, Liu Y, Deng M, Ke D, Wang Q, Wang Y, Wang JZ. A novel small-molecule PROTAC selectively promotes tau clearance to improve cognitive functions in Alzheimer-like models. Theranostics 2021; 11:5279-5295. [PMID: 33859747 PMCID: PMC8039949 DOI: 10.7150/thno.55680] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/18/2021] [Indexed: 12/21/2022] Open
Abstract
Intracellular accumulation of tau is a hallmark pathology in Alzheimer disease (AD) and the related tauopathies, thus targeting tau could be promising for drug development. Proteolysis Targeting Chimera (PROTAC) is a novel drug discovery strategy for selective protein degradation from within cells. Methods: A novel small-molecule PROTAC, named as C004019 with a molecular mass of 1,035.29 dalton, was designed to simultaneously recruite tau and E3-ligase (Vhl) and thus to selectively enhance ubiquitination and proteolysis of tau proteins. Western blotting, immunofluoresence and immunohistochemical staining were employed to verify the effects of C004019 in cell models (HEK293 and SH-SY5Y) and mouse models (hTau-transgenic and 3xTg-AD), respectively. The cognitive capacity of the mice was assessed by a suite of behavior experiments. Electrophysiology and Golgi staining were used to evaluate the synaptic plasticity. Results: C004019 induced a robust tau clearance via promoting its ubiquitination-proteasome-dependent proteolysis in HEK293 cells with stable or transient overexpression of human tau (hTau), and in SH-SY5Y that constitutively overexpress hTau. Furthermore, intracerebral ventricular infusion of C004019 induced a robust tau clearance in vivo. Most importantly, both single-dose and multiple-doses (once per 6 days for a total 5 times) subcutaneous administration of C004019 remarkably decreased tau levels in the brains of wild-type, hTau-transgenic and 3xTg-AD mice with improvement of synaptic and cognitive functions. Conclusions: The PROTAC (C004019) created in the current study can selectively and efficiently promote tau clearance both in vitro and in vivo, which provides a promising drug candidate for AD and the related tauopathies.
Collapse
Affiliation(s)
- Weijin Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qiuzhi Zhou
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Jiang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shihong Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jinwang Ye
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jie Zheng
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xin Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yanchao Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Minmin Deng
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dan Ke
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qun Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yipeng Wang
- Neurosmart Therapeutics Co., Ltd., Room 5013, Unit 1, Buiilding 7, Basheng road 160, Shanghai 200131, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226000, China
| |
Collapse
|
220
|
Amin S, Alam MM, Akhter M, Najmi AK, Siddiqui N, Husain A, Shaquiquzzaman M. A review on synthetic procedures and applications of phosphorus oxychloride (POCl 3) in the last biennial period (2018–19). PHOSPHORUS SULFUR 2021. [DOI: 10.1080/10426507.2020.1831499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Shaista Amin
- Drug Design & Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - M. Mumtaz Alam
- Drug Design & Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Mymoona Akhter
- Drug Design & Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - A. K. Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Nadeem Siddiqui
- Drug Design & Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Asif Husain
- Drug Design & Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - M. Shaquiquzzaman
- Drug Design & Medicinal Chemistry Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|
221
|
Wan Yaacob WMH, Long I, Zakaria R, Othman Z. Tualang honey and its methanolic fraction ameliorate lipopolysaccharide-induced oxidative stress, amyloid deposition and neuronal loss of the rat hippocampus. ADVANCES IN TRADITIONAL MEDICINE 2021. [DOI: 10.1007/s13596-020-00449-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
222
|
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: 1.0] [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
|
223
|
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: 32] [Impact Index Per Article: 10.7] [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
|
224
|
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: 1.3] [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
|
225
|
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: 0.7] [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
|
226
|
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: 35] [Impact Index Per Article: 11.7] [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
|
227
|
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: 2.0] [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
|
228
|
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: 2.3] [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
|
229
|
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: 4.0] [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
|
230
|
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
|
231
|
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: 23] [Impact Index Per Article: 7.7] [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
|
232
|
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: 11] [Impact Index Per Article: 3.7] [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
|
233
|
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: 12] [Impact Index Per Article: 4.0] [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
|
234
|
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: 42] [Impact Index Per Article: 14.0] [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
|
235
|
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
|
236
|
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: 2.8] [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
|
237
|
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: 1.5] [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
|
238
|
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: 0.5] [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
|
239
|
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: 1.0] [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
|
240
|
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: 1.3] [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
|
241
|
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: 0.8] [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
|
242
|
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: 4.3] [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
|
243
|
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: 0.3] [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
|
244
|
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: 97] [Impact Index Per Article: 24.3] [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
|
245
|
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: 0.8] [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
|
246
|
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: 2.0] [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
|
247
|
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: 10] [Impact Index Per Article: 2.5] [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
|
248
|
Panchal K, Tiwari AK. Miro (Mitochondrial Rho GTPase), a key player of mitochondrial axonal transport and mitochondrial dynamics in neurodegenerative diseases. Mitochondrion 2020; 56:118-135. [PMID: 33127590 DOI: 10.1016/j.mito.2020.10.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
249
|
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: 0.3] [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
|
250
|
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: 13] [Impact Index Per Article: 3.3] [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
|