1
|
Ji CH, Shin DH, Son YH, Kam TE. Sparse Graph Representation Learning Based on Reinforcement Learning for Personalized Mild Cognitive Impairment (MCI) Diagnosis. IEEE J Biomed Health Inform 2024; 28:4842-4853. [PMID: 38683720 DOI: 10.1109/jbhi.2024.3393625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Resting-state functional magnetic resonance imaging (rs-fMRI) has gained attention as a reliable technique for investigating the intrinsic function patterns of the brain. It facilitates the extraction of functional connectivity networks (FCNs) that capture synchronized activity patterns among regions of interest (ROIs). Analyzing FCNs enables the identification of distinctive connectivity patterns associated with mild cognitive impairment (MCI). For MCI diagnosis, various sparse representation techniques have been introduced, including statistical- and deep learning-based methods. However, these methods face limitations due to their reliance on supervised learning schemes, which restrict the exploration necessary for probing novel solutions. To overcome such limitation, prior work has incorporated reinforcement learning (RL) to dynamically select ROIs, but effective exploration remains challenging due to the vast search space during training. To tackle this issue, in this study, we propose an advanced RL-based framework that utilizes a divide-and-conquer approach to decompose the FCN construction task into smaller sub-problems in a subject-specific manner, enabling efficient exploration under each sub-problem condition. Additionally, we leverage the learned value function to determine the sparsity level of FCNs, considering individual characteristics of FCNs. We validate the effectiveness of our proposed framework by demonstrating its superior performance in MCI diagnosis on publicly available cohort datasets.
Collapse
|
2
|
Suliman M, Al-Hawary SIS, Al-Dolaimy F, Hjazi A, Almalki SG, Alkhafaji AT, Alawadi AH, Alsaalamy A, Bijlwan S, Mustafa YF. Inflammatory diseases: Function of LncRNAs in their emergence and the role of mesenchymal stem cell secretome in their treatment. Pathol Res Pract 2023; 249:154758. [PMID: 37660657 DOI: 10.1016/j.prp.2023.154758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 09/05/2023]
Abstract
One of the best treatments for inflammatory diseases such as COVID-19, respiratory diseases and brain diseases is treatment with stem cells. Here we investigate the effect of stem cell therapy in the treatment of brain diseases.Preclinical studies have shown promising results, including improved functional recovery and tissue repair in animal models of neurodegenerative diseases, strokes,and traumatic brain injuries. However,ethical implications, safety concerns, and regulatory frameworks necessitate thorough evaluation before transitioning to clinical applications. Additionally, the complex nature of the brain and its intricate cellular environment present unique obstacles that must be overcome to ensure the successful integration and functionality of genetically engineered MSCs. The careful navigation of this path will determine whether the application of genetically engineered MSCs in brain tissue regeneration ultimately lives up to the hype surrounding it.
Collapse
Affiliation(s)
- Muath Suliman
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | | | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia.
| | - Sami G Almalki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, Saudi Arabia
| | | | - Ahmed Hussien Alawadi
- College of technical engineering, the Islamic University, Najaf, Iraq; College of technical engineering, the Islamic University of Al Diwaniyah, Iraq; College of technical engineering, the Islamic University of Babylon, Iraq
| | - Ali Alsaalamy
- College of technical engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna, Iraq
| | - Sheela Bijlwan
- Uttaranchal School of Computing Sciences, Uttaranchal University, Dehradun, India
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| |
Collapse
|
3
|
Abstract
Reviewing the research presented in this article, it is evident that from an epidemiological perspective, it is important to evaluate the extent to which findings of sex and gender differences in Alzheimer's dementia (AD) are due to differences in longevity, survival bias, and comorbidities. Medical, genetic, psychosocial, and behavioral factors, in addition to hormonal factors, can differentially affect the risk and progression of AD in women versus men. Further, evaluation of sex differences in AD progression and the trajectory of change in cognitive function, neuroimaging, cerebrospinal fluid (CSF), and blood-based biomarkers of AD is needed. Finally, identifying sex differences in AD biomarkers and change across the lifespan is critical for the planning of prevention trials to reduce the risk of developing AD.
Collapse
Affiliation(s)
- Neelum T Aggarwal
- Department of Neurological Sciences, Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 West Harrison Street, Suite 1000, Chicago, IL 60612, USA.
| | - Michelle M Mielke
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
4
|
Ozkat GY, Yildiz I. In Silico Studies to Develop New GSK3β Inhibitors Effective in Alzheimer's Disease. LETT DRUG DES DISCOV 2022. [DOI: 10.2174/1570180819666220210100813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Alzheimer's disease affects a large part of the world population by prolonging the human life span and becoming an economic burden in the health system. Therefore, its treatment becomes more and more important every day. With the insufficiency of existing drug molecules, new drug targets started to be emphasized. The most important of these is the Glycogen Synthase Kinase 3β enzyme, thought to be of key importance in Tau hyperphosphorylation and Amyloid β accumulation mechanisms.
Objective:
In this research, computational studies were conducted to develop a new GSK3β enzyme inhibitor.
Method:
Leading compounds suitable for pharmacophore models obtained by the 3D QSAR method were scanned in databases. In silico ADME/Tox analyses were performed on the obtained molecules.
Results:
Although the three molecules (ENA99104, CNR13756, TIM405938) had strong Dock Scores (42.869, 53.344, and 41.119, respectively) in molecular docking calculations, only the CNR13756 molecule was found successful according to molecular dynamics simulations.
Conclusion:
All computational studies have revealed that the CNR13756 molecule can exhibit a therapeutic scaffold property, thus obtaining a selective GSK3β inhibitor with minimal side effects.
Collapse
Affiliation(s)
- Gozde Yalcin Ozkat
- Biotechnology Institute, Ankara University, Ankara 06135 Turkey
- Bioengineering Department, Faculty of Engineering and Architecture, Recep Tayyip Erdogan University, Rize, Turkey
| | - Ilkay Yildiz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, Ankara 06110, Turkey
| |
Collapse
|
5
|
Amin M, Tang S, Shalamanova L, Taylor RL, Wylie S, Abdullah BM, Whitehead KA. Polyamine biomarkers as indicators of human disease. Biomarkers 2021; 26:77-94. [PMID: 33439737 DOI: 10.1080/1354750x.2021.1875506] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The significant increase of periodontitis, chronic kidney disease (CKD), Alzheimer's disease and cancer can be attributed to an ageing population. Each disease produces a range of biomarkers that can be indicative of disease onset and progression. Biomarkers are defined as cellular (intra/extracellular components and whole cells), biochemical (metabolites, ions and toxins) or molecular (nucleic acids, proteins and lipids) alterations which are measurable in biological media such as human tissues, cells or fluids. An interesting group of biomarkers that merit further investigation are the polyamines. Polyamines are a group of molecules consisting of cadaverine, putrescine, spermine and spermidine and have been implicated in the development of a range of systemic diseases, in part due to their production in periodontitis. Cadaverine and putrescine within the periodontal environment have demonstrated cell signalling interfering abilities, by way of leukocyte migration disruption. The polyamines spermine and spermidine in tumour cells have been shown to inhibit cellular apoptosis, effectively prolonging tumorigenesis and continuation of cancer within the host. Polyamine degradation products such as acrolein have been shown to exacerbate renal damage in CKD patients. Thus, the use of such molecules has merit to be utilized in the early indication of such diseases in patients.
Collapse
Affiliation(s)
- Mohsin Amin
- Microbiology at Interfaces, Manchester Metropolitan University, Manchester, UK.,Department of Engineering and Technology, Built Environment, Liverpool John Moores University, Liverpool, UK
| | - Shiying Tang
- Microbiology at Interfaces, Manchester Metropolitan University, Manchester, UK.,Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Liliana Shalamanova
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Rebecca L Taylor
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Stephen Wylie
- Department of Engineering and Technology, Civil Engineering, Liverpool John Moores University, Liverpool, UK
| | - Badr M Abdullah
- Department of Engineering and Technology, Built Environment, Liverpool John Moores University, Liverpool, UK
| | - Kathryn A Whitehead
- Microbiology at Interfaces, Manchester Metropolitan University, Manchester, UK.,Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| |
Collapse
|
6
|
Effect of Treadmill Exercise and Trans-Cinnamaldehyde against d-Galactose- and Aluminum Chloride-Induced Cognitive Dysfunction in Mice. Brain Sci 2020; 10:brainsci10110793. [PMID: 33138104 PMCID: PMC7693345 DOI: 10.3390/brainsci10110793] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 10/25/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023] Open
Abstract
Mild cognitive impairment (MCI) generally refers to impairment in cognition above that which accompanies the normal age-related cognitive decline and has attracted attention in recent years. Trans-cinnamaldehyde (TCA), which is isolated from cinnamon, has anti-inflammatory and antioxidant properties. Treadmill exercise also has diverse positive effects. The purpose of this study was to investigate the combination effects of TCA and treadmill exercise on learning and memory in a cognitive impairment mouse induced by a combination of d-galactose (d-gal) and aluminum chloride (AlCl3). We found that exercise and TCA attenuated cognitive impairment in mice with induced MCI. This effect was further increased by costimulation of exercise and TCA. To clarify the mechanisms of the positive effects of TCA and exercise, we analyzed the nuclear factor erythroid 2-related factor (Nrf2) and related signaling pathways. We found that TCA and exercise upregulated Nrf2, NAD(P)H dehydrogenase quinone 1 (NQO-1), heme oxygenase 1 (HO-1), and superoxide dismutase 1 (SOD-1); this suggests that TCA and exercise attenuate cognitive dysfunction by reducing oxidative stress. We also found that Nrf2-related signaling pathways, i.e., the AMP-activated protein kinase (AMPK)/Nrf2 and SIRT1/PGC-1a/Nrf2-ARE pathways, exerted antioxidant effects. Together, these results suggest that costimulation with TCA and exercise may be a therapeutic candidate for mild cognitive impairment.
Collapse
|
7
|
Lesman-Segev OH, Edwards L, Rabinovici GD. Chronic Traumatic Encephalopathy: A Comparison with Alzheimer's Disease and Frontotemporal Dementia. Semin Neurol 2020; 40:394-410. [PMID: 32820492 DOI: 10.1055/s-0040-1715134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The clinical diagnosis of chronic traumatic encephalopathy (CTE) is challenging due to heterogeneous clinical presentations and overlap with other neurodegenerative dementias. Depending on the clinical presentation, the differential diagnosis of CTE includes Alzheimer's disease (AD), behavioral variant frontotemporal dementia (bvFTD), Parkinson's disease, amyotrophic lateral sclerosis, primary mood disorders, posttraumatic stress disorder, and psychotic disorders. The aim of this article is to compare the clinical aspects, genetics, fluid biomarkers, imaging, treatment, and pathology of CTE to those of AD and bvFTD. A detailed clinical evaluation, neurocognitive assessment, and structural brain imaging can inform the differential diagnosis, while molecular biomarkers can help exclude underlying AD pathology. Prospective studies that include clinicopathological correlations are needed to establish tools that can more accurately determine the cause of neuropsychiatric decline in patients at risk for CTE.
Collapse
Affiliation(s)
- Orit H Lesman-Segev
- Department of Neurology, University of California San Francisco, San Francisco, California
| | - Lauren Edwards
- Department of Neurology, University of California San Francisco, San Francisco, California
| | - Gil D Rabinovici
- Department of Neurology, University of California San Francisco, San Francisco, California.,Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California.,Weill Neuroscience Institute, University of California San Francisco, San Francisco, California
| |
Collapse
|
8
|
Gavrilova SI, Alvarez A. Cerebrolysin in the therapy of mild cognitive impairment and dementia due to Alzheimer's disease: 30 years of clinical use. Med Res Rev 2020; 41:2775-2803. [PMID: 32808294 DOI: 10.1002/med.21722] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/01/2020] [Accepted: 08/05/2020] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is the most common neurocognitive disorder and a global health problem. The prevalence of AD is growing dramatically, especially in low- and middle-income countries, and will reach 131.5 million cases worldwide by 2050. Therefore, developing a disease-modifying therapy capable of delaying or even preventing the onset and progression of AD has become a world priority, and is an unmet need. The pathogenesis of AD, considered as the result of an imbalance between resilience and risk factors, begins many years before the typical clinical picture develops and involves multiple pathophysiological mechanisms. Since the pathophysiology of AD is multifactorial, it is not surprising that all attempts done to modify the disease course with drugs directed towards a single therapeutic target have been unsuccessful. Thus, combined modality therapy, using multiple drugs with a single mechanism of action or multi-target drugs, appears as the most promising strategy for both effective AD therapy and prevention. Cerebrolysin, acting as a multitarget peptidergic drug with a neurotrophic mode of action, exerts long-lasting therapeutic effects on AD that could reflect its potential utility for disease modification. Clinical trials demonstrated that Cerebrolysin is safe and efficacious in the treatment of AD, and may enhance and prolong the efficacy of cholinergic drugs, particularly in moderate to advanced AD patients. In this review, we summarize advances of therapeutic relevance in the pathogenesis and the biomarkers of AD, paying special attention to neurotrophic factors, and present results of preclinical and clinical investigations with Cerebrolysin in AD.
Collapse
Affiliation(s)
- Svetlana I Gavrilova
- Department of Geriatric Psychiatry, Cognitive Disorders and Alzheimer's Disease Unit, Mental Health Research Center, Moscow, Russia
| | - Anton Alvarez
- Department of Neuropsychiatry, Medinova Institute of Neurosciences, Clinica RehaSalud, A Coruña, Spain.,Clinical Research Department, QPS Holdings, A Coruña, Spain
| |
Collapse
|
9
|
Moss DE. Improving Anti-Neurodegenerative Benefits of Acetylcholinesterase Inhibitors in Alzheimer's Disease: Are Irreversible Inhibitors the Future? Int J Mol Sci 2020; 21:E3438. [PMID: 32414155 PMCID: PMC7279429 DOI: 10.3390/ijms21103438] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/01/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023] Open
Abstract
Decades of research have produced no effective method to prevent, delay the onset, or slow the progression of Alzheimer's disease (AD). In contrast to these failures, acetylcholinesterase (AChE, EC 3.1.1.7) inhibitors slow the clinical progression of the disease and randomized, placebo-controlled trials in prodromal and mild to moderate AD patients have shown AChE inhibitor anti-neurodegenerative benefits in the cortex, hippocampus, and basal forebrain. CNS neurodegeneration and atrophy are now recognized as biomarkers of AD according to the National Institute on Aging-Alzheimer's Association (NIA-AA) criteria and recent evidence shows that these markers are among the earliest signs of prodromal AD, before the appearance of amyloid. The current AChE inhibitors (donepezil, rivastigmine, and galantamine) have short-acting mechanisms of action that result in dose-limiting toxicity and inadequate efficacy. Irreversible AChE inhibitors, with a long-acting mechanism of action, are inherently CNS selective and can more than double CNS AChE inhibition possible with short-acting inhibitors. Irreversible AChE inhibitors open the door to high-level CNS AChE inhibition and improved anti-neurodegenerative benefits that may be an important part of future treatments to more effectively prevent, delay the onset, or slow the progression of AD.
Collapse
Affiliation(s)
- Donald E Moss
- Department of Psychology, University of Texas at El Paso, El Paso, TX 79968, USA
| |
Collapse
|
10
|
NAVIPOUR E, NEAMATSHAHI M, BARABADI Z, NEAMATSHAHI M, KEYKHOSRAVI A. Epidemiology and Risk Factors of Alzheimer's Disease in Iran: A Systematic Review. IRANIAN JOURNAL OF PUBLIC HEALTH 2019; 48:2133-2139. [PMID: 31993381 PMCID: PMC6974866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 06/09/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Alzheimer's disease is a chronic disease characterized by a progressive decline in mental abilities and quality of life alongside behavioral abnormalities associated with high economic burden. The purpose of this study was to investigate epidemiology and risk factors of Alzheimer's disease in Iran. METHODS In this systematic review study, both Persian and English-language databases including Medline, Google Scholar, PubMed, web of science and Magiran were searched using following keywords: epidemiology, Alzheimer, dementia and Iran without time limit up to 2017. Thirty articles abstract out of 50 studies related to this topics, were reviewed. Of which 12 full text entered into the quality assessment process and finally, four articles were selected for inclusion in this study and their results was extracted. RESULTS The total sample size of the 4 selected studies was 2781. The prevalence of Alzheimer's disease in the current study was estimated to be 2.3% in the population of 67-78 years old. Age, genetics, depression and hypertension were determined as the risk factors for Alzheimer's disease, while daily listening to music, meeting weekly with friends and daily intake of vitamin E were considered as the factors with protective role in this disease. CONCLUSION Alzheimer's disease is one of the main causes of functional dependence and mortality in the elderly people. Lifestyle changes and multiple mental activities in elderly increases the cognitive ability of these population, which will reduce direct and indirect costs of this disease.
Collapse
Affiliation(s)
- Elham NAVIPOUR
- Department of Biostatistics, Faculty of Paramedicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Mahbobeh NEAMATSHAHI
- Department of Community Medicine, Faculty of Medicine, Research Center Social Determinants Health, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Zahra BARABADI
- Department of Physiology and Pharmacology, School of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Mohammad NEAMATSHAHI
- Department of Anesthesiology, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Aghilallah KEYKHOSRAVI
- Department of Pediatrics, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| |
Collapse
|
11
|
Vyhnálek M, Marková H, Laczó J, De Beni R, Di Nuovo S. Assessment of Memory Impairment in Early Diagnosis of Alzheimer's Disease. Curr Alzheimer Res 2019; 16:975-985. [PMID: 31724515 DOI: 10.2174/1567205016666191113125303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 11/02/2019] [Accepted: 11/05/2019] [Indexed: 11/22/2022]
Abstract
Memory impairment has been considered as one of the earliest clinical hallmarks of Alzheimer's disease. This paper summarizes recent progress in the assessment of memory impairment in predementia stages. New promising approaches of memory assessment include evaluation of longitudinal cognitive changes, assessment of long-term memory loss, evaluation of subjective cognitive concerns and testing of other memory modalities, such as spatial memory. In addition, we describe new challenging memory tests based on memory binding paradigms that have been recently developed and are currently being validated.
Collapse
Affiliation(s)
- Martin Vyhnálek
- Memory Clinic, Department of Neurology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Hana Marková
- Memory Clinic, Department of Neurology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Jan Laczó
- Memory Clinic, Department of Neurology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | | | - Santo Di Nuovo
- Department of Education, University of Catania, Catania, Italy
| |
Collapse
|
12
|
Chakari-Khiavi F, Dolati S, Chakari-Khiavi A, Abbaszadeh H, Aghebati-Maleki L, Pourlak T, Mehdizadeh A, Yousefi M. Prospects for the application of mesenchymal stem cells in Alzheimer's disease treatment. Life Sci 2019; 231:116564. [PMID: 31202840 DOI: 10.1016/j.lfs.2019.116564] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 06/11/2019] [Accepted: 06/11/2019] [Indexed: 01/09/2023]
Abstract
Alzheimer's disease (AD) as a dementia and neurodegenerative disease, is mostly prevalent among people more than 65 years. AD is mostly manifested in the form of degraded mental function, such as losing memory and impaired cognitive function. Due to inefficiency of traditional pharmacological therapeutic approaches with no long-term cure, cell therapy can be considered as a capable approach in AD management. Therapies based on mesenchymal stem cells (MSCs) have provided hopeful results in experimental models regarding several disorders. MSCs enhance the levels of functional recoveries in pathologic experimental models of central nervous system (CNS) and are being investigated in clinical trials in neurological disorders. However, there is limited knowledge on the protective capabilities of MSCs in AD management. Almost, several experiments have suggested positive effects of MSCs and helped to better understand of AD-related dementia mechanism. MSCs have the potential to be used in AD treatment through amyloid-β peptide (AB), Tau protein and cholinergic system. This review aimed to clarify the promising perspective of MSCs in the context of AD.
Collapse
Affiliation(s)
- Forough Chakari-Khiavi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanam Dolati
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Aging Research Institute, Tabriz University of Medical Sciences Tabriz, Iran
| | - Aref Chakari-Khiavi
- Aging Research Institute, Tabriz University of Medical Sciences Tabriz, Iran
| | - Hossein Abbaszadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Tannaz Pourlak
- Aging Research Institute, Tabriz University of Medical Sciences Tabriz, Iran
| | - Amir Mehdizadeh
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Aging Research Institute, Tabriz University of Medical Sciences Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran..
| |
Collapse
|
13
|
de Castro AA, Soares FV, Pereira AF, Polisel DA, Caetano MS, Leal DHS, da Cunha EFF, Nepovimova E, Kuca K, Ramalho TC. Non-conventional compounds with potential therapeutic effects against Alzheimer’s disease. Expert Rev Neurother 2019; 19:375-395. [DOI: 10.1080/14737175.2019.1608823] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Alexandre A. de Castro
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras, Brazil
| | - Flávia V. Soares
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras, Brazil
| | - Ander F. Pereira
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras, Brazil
| | - Daniel A. Polisel
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras, Brazil
| | - Melissa S. Caetano
- Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Daniel H. S. Leal
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras, Brazil
- Department of Health Sciences, Federal University of Espírito Santo, São Mateus, Brazil
| | - Elaine F. F. da Cunha
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras, Brazil
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Teodorico C. Ramalho
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras, Brazil
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
14
|
Rabinovici GD, Gatsonis C, Apgar C, Chaudhary K, Gareen I, Hanna L, Hendrix J, Hillner BE, Olson C, Lesman-Segev OH, Romanoff J, Siegel BA, Whitmer RA, Carrillo MC. Association of Amyloid Positron Emission Tomography With Subsequent Change in Clinical Management Among Medicare Beneficiaries With Mild Cognitive Impairment or Dementia. JAMA 2019; 321:1286-1294. [PMID: 30938796 PMCID: PMC6450276 DOI: 10.1001/jama.2019.2000] [Citation(s) in RCA: 351] [Impact Index Per Article: 70.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
IMPORTANCE Amyloid positron emission tomography (PET) detects amyloid plaques in the brain, a core neuropathological feature of Alzheimer disease. OBJECTIVE To determine if amyloid PET is associated with subsequent changes in the management of patients with mild cognitive impairment (MCI) or dementia of uncertain etiology. DESIGN, SETTING, AND PARTICIPANTS The Imaging Dementia-Evidence for Amyloid Scanning (IDEAS) study was a single-group, multisite longitudinal study that assessed the association between amyloid PET and subsequent changes in clinical management for Medicare beneficiaries with MCI or dementia. Participants were required to meet published appropriate use criteria stating that etiology of cognitive impairment was unknown, Alzheimer disease was a diagnostic consideration, and knowledge of PET results was expected to change diagnosis and management. A total of 946 dementia specialists at 595 US sites enrolled 16 008 patients between February 2016 and September 2017. Patients were followed up through January 2018. Dementia specialists documented their diagnosis and management plan before PET and again 90 (±30) days after PET. EXPOSURES Participants underwent amyloid PET at 343 imaging centers. MAIN OUTCOMES AND MEASURES The primary end point was change in management between the pre- and post-PET visits, as assessed by a composite outcome that included Alzheimer disease drug therapy, other drug therapy, and counseling about safety and future planning. The study was powered to detect a 30% or greater change in the MCI and dementia groups. One of 2 secondary end points is reported: the proportion of changes in diagnosis (from Alzheimer disease to non-Alzheimer disease and vice versa) between pre- and post-PET visits. RESULTS Among 16 008 registered participants, 11 409 (71.3%) completed study procedures and were included in the analysis (median age, 75 years [interquartile range, 71-80]; 50.9% women; 60.5% with MCI). Amyloid PET results were positive in 3817 patients with MCI (55.3%) and 3154 patients with dementia (70.1%). The composite end point changed in 4159 of 6905 patients with MCI (60.2% [95% CI, 59.1%-61.4%]) and 2859 of 4504 patients with dementia (63.5% [95% CI, 62.1%-64.9%]), significantly exceeding the 30% threshold in each group (P < .001, 1-sided). The etiologic diagnosis changed from Alzheimer disease to non-Alzheimer disease in 2860 of 11 409 patients (25.1% [95% CI, 24.3%-25.9%]) and from non-Alzheimer disease to Alzheimer disease in 1201 of 11 409 (10.5% [95% CI, 10.0%-11.1%]). CONCLUSIONS AND RELEVANCE Among Medicare beneficiaries with MCI or dementia of uncertain etiology evaluated by dementia specialists, the use of amyloid PET was associated with changes in clinical management within 90 days. Further research is needed to determine whether amyloid PET is associated with improved clinical outcomes. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02420756.
Collapse
Affiliation(s)
- Gil D. Rabinovici
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
- Associate Editor, JAMA Neurology
| | - Constantine Gatsonis
- Center for Statistical Sciences, Brown University School of Public Health, Providence, Rhode Island
- Department of Biostatistics, Brown University School of Public Health, Providence, Rhode Island
| | | | - Kiran Chaudhary
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Ilana Gareen
- Center for Statistical Sciences, Brown University School of Public Health, Providence, Rhode Island
- Department of Epidemiology, Brown University School of Public Health, Providence, Rhode Island
| | - Lucy Hanna
- Center for Statistical Sciences, Brown University School of Public Health, Providence, Rhode Island
| | | | - Bruce E. Hillner
- Department of Medicine, Virginia Commonwealth University, Richmond
| | | | - Orit H. Lesman-Segev
- Memory and Aging Center, Department of Neurology, University of California, San Francisco
| | - Justin Romanoff
- Center for Statistical Sciences, Brown University School of Public Health, Providence, Rhode Island
| | - Barry A. Siegel
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Rachel A. Whitmer
- Division of Research, Kaiser Permanente, Oakland, California
- Department of Public Health Sciences, University of California, Davis
| | | |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW Alzheimer disease (AD) is the most common cause of late-onset dementia. This article describes the epidemiology, genetic and environmental risk factors, clinical diagnosis, biomarkers, and treatment of late-onset AD, defined by age of onset of 65 years or older. RECENT FINDINGS An estimated 5.7 million Americans are living with AD dementia, with the number of affected individuals growing rapidly because of an aging population. Vascular risk factors, sleep disorders, and traumatic brain injury are associated with an increased risk of AD, while increased cognitive and physical activity throughout the lifespan reduce the risk of disease. The primary genetic risk factor for late-onset AD is the apolipoprotein E (APOE) ε4 allele. AD typically presents with early and prominent episodic memory loss, although this clinical syndrome is neither sensitive nor specific for underlying AD neuropathology. Emerging CSF and imaging biomarkers can now detect the key neuropathologic features of the disease (amyloid plaques, neurofibrillary tangles, and neurodegeneration) in living people, allowing for characterization of patients based on biological measures. A comprehensive treatment plan for AD includes use of symptomatic medications, optimal treatment of comorbid conditions and neuropsychiatric symptoms, counseling about safety and future planning, and referrals to community resources. SUMMARY AD is very common in older neurologic patients. Neurologists should set the standard for the diagnosis and care of patients with AD and should be familiar with emerging biomarkers that have transformed AD research and are primed to enter the clinical arena.
Collapse
|
16
|
Ginkgo biloba Extract Decreases Scopolamine-Induced Congophilic Amyloid Plaques Accumulation in Male Rat's Brain. Jundishapur J Nat Pharm Prod 2018. [DOI: 10.5812/jjnpp.69143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
17
|
Deering Brose R, Lehrmann E, Zhang Y, Reeves RH, Smith KD, Mattson MP. Hydroxyurea attenuates oxidative, metabolic, and excitotoxic stress in rat hippocampal neurons and improves spatial memory in a mouse model of Alzheimer's disease. Neurobiol Aging 2018; 72:121-133. [PMID: 30245242 PMCID: PMC6215724 DOI: 10.1016/j.neurobiolaging.2018.08.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 08/17/2018] [Accepted: 08/23/2018] [Indexed: 10/28/2022]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder characterized by accumulation of amyloid β-peptide (Aβ) plaques in the brain and decreased cognitive function leading to dementia. We tested if hydroxyurea (HU), a ribonucleotide reductase inhibitor known to activate adaptive cellular stress responses and ameliorate abnormalities associated with several genetic disorders, could protect rat hippocampal neurons against oxidative-, excitatory-, mitochondrial-, and Aβ-induced stress and if HU treatment could improve learning and memory in the APP/PS1 mouse model of AD. HU treatment attenuated the loss of cell viability induced by treatment of hippocampal neurons with hydrogen peroxide, glutamate, rotenone, and Aβ1-42. HU treatment attenuated reductions of mitochondrial reserve capacity, maximal respiration, and cellular adenosine triphosphate content induced by hydrogen peroxide treatment. In vivo, treatment of APP/PS1 mice with HU (45 mg/kg/d) improved spatial memory performance in the hippocampus-dependent Morris water maze task without reducing Aβ levels. HU provides neuroprotection against toxic insults including Aβ, improves mitochondrial bioenergetics, and improves spatial memory in an AD mouse model. HU may offer a new therapeutic approach to delay cognitive decline in AD.
Collapse
Affiliation(s)
- Rebecca Deering Brose
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21025, USA , ,
- Laboratory of Neurosciences, National Institutes of Health, 251 Bayview Blvd, Suite 100, Baltimore, MD 21224, USA
| | - Elin Lehrmann
- Laboratory of Genetics and Genomics, National Institute of Aging, National Institutes of Health, 251 Bayview Blvd, Suite 100, Baltimore, MD 21224, USA , ,
| | - Yongqing Zhang
- Laboratory of Genetics and Genomics, National Institute of Aging, National Institutes of Health, 251 Bayview Blvd, Suite 100, Baltimore, MD 21224, USA , ,
| | - Roger H. Reeves
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21025, USA , ,
| | - Kirby D. Smith
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21025, USA , ,
| | - Mark P. Mattson
- Laboratory of Neurosciences, National Institutes of Health, 251 Bayview Blvd, Suite 100, Baltimore, MD 21224, USA
| |
Collapse
|
18
|
Elfakhri KH, Duong QV, Langley C, Depaula A, Mousa YM, Lebeouf T, Cain C, Kaddoumi A. Characterization of Hit Compounds Identified from High-throughput Screening for their Effect on Blood-brain Barrier Integrity and Amyloid-β Clearance: In Vitro and In Vivo Studies. Neuroscience 2018; 379:269-280. [PMID: 29596966 PMCID: PMC5924725 DOI: 10.1016/j.neuroscience.2018.03.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/05/2018] [Accepted: 03/19/2018] [Indexed: 11/15/2022]
Abstract
In Alzheimer's disease (AD) the blood-brain barrier (BBB) is compromised, thus therapeutic targeting of the BBB to enhance its integrity and function could be a unique approach to treat, slow or hold the progression of AD. Recently, we have developed an in vitro high-throughput screening assay to screen for compounds that increase the integrity of a cell-based BBB model. Results from primary screen identified multiple hit compounds that enhanced the monolayer integrity. Herein, further characterization of selected hit compounds, namely 8-bromoguanosine cyclic monophosphate, JW74, 1,10-phenanthroline monohydrate, SB216763 and α-tocopherol was performed. Compounds were subjected to concentration-dependent studies to determine their EC50 and potency to enhance the cell-based model integrity by the Lucifer Yellow permeability and amyloid-beta (Aβ) transport across the monolayer. The compounds demonstrated different EC50s to enhance the monolayer integrity ranging from 0.4 to 12.8 µM, and different effect on enhancing Aβ transport with highest transport observed for α-tocopherol (2.2-fold increase). Such effects were associated with increased levels of tight junction proteins such as claudin-5 and/or ZO-1, and Aβ major transport proteins LRP1 and P-glycoprotein. In vivo studies for α-tocopherol were performed in AD mouse model; consistent with the in vitro results α-tocopherol significantly increased BBB integrity measured by IgG extravasation, and reduced brain Aβ levels. In conclusion, findings support our developed cell-based BBB model as a functional predictive in vivo tool to select hit compounds, and suggest that enhancing BBB tightness and function has the potential to reduce Aβ pathology associated with AD.
Collapse
Affiliation(s)
- Khaled H Elfakhri
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Quoc-Viet Duong
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Courtney Langley
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Ashley Depaula
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Youssef M Mousa
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Trista Lebeouf
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Courtney Cain
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Amal Kaddoumi
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA.
| |
Collapse
|
19
|
Khoury R, Patel K, Gold J, Hinds S, Grossberg GT. Recent Progress in the Pharmacotherapy of Alzheimer's Disease. Drugs Aging 2018; 34:811-820. [PMID: 29116600 DOI: 10.1007/s40266-017-0499-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease is the most common major neurocognitive disorder with substantial social and economic impacts. This article is an update on current pharmacotherapy, advancements in biomarker use, and drugs in the pipeline for this disease. To date, no new drug has qualified to be added to the current therapeutic arsenal comprising cholinesterase inhibitors and the NMDA receptor antagonist memantine. Drugs in the pipeline include symptomatic therapies that are neurotransmitter-based, but mostly disease-modifying therapies. The latter have yielded disappointing results by focusing mainly on the two pathophysiological hallmarks of Alzheimer's disease: Aβ amyloid deposits and tau protein aggregates forming neurofibrillary tangles. These unsuccessful trials may have resulted from studying these drugs 'too late' relative to Alzheimer's disease onset, in addition to focusing only on the amyloid cascade. In fact, Alzheimer's disease is a complex multifactorial disease. Combining different biomarkers might enhance our ability to identify those patients most at risk of developing the disease, and better predict their conversion rates. Furthermore, adopting an integrative treatment approach by targeting additional pathophysiological pathways in Alzheimer's disease such as inflammation and oxidative stress could be the key to better outcomes in Alzheimer's disease pharmacotherapy research.
Collapse
Affiliation(s)
- Rita Khoury
- Division of Geriatric Psychiatry, St. Louis University School of Medicine, 1438 S Grand Blvd, St. Louis, MO, 63104, USA.
| | - Kush Patel
- Division of Geriatric Psychiatry, St. Louis University School of Medicine, 1438 S Grand Blvd, St. Louis, MO, 63104, USA
| | - Jake Gold
- Division of Geriatric Psychiatry, St. Louis University School of Medicine, 1438 S Grand Blvd, St. Louis, MO, 63104, USA
| | - Stephanie Hinds
- Division of Geriatric Psychiatry, St. Louis University School of Medicine, 1438 S Grand Blvd, St. Louis, MO, 63104, USA
| | - George T Grossberg
- Division of Geriatric Psychiatry, St. Louis University School of Medicine, 1438 S Grand Blvd, St. Louis, MO, 63104, USA
| |
Collapse
|
20
|
Hernández-Zimbrón LF, Perez-Hernández M, Torres-Romero A, Gorostieta-Salas E, Gonzalez-Salinas R, Gulias-Cañizo R, Quiroz-Mercado H, Zenteno E. Markers of Alzheimer's Disease in Primary Visual Cortex in Normal Aging in Mice. BIOMED RESEARCH INTERNATIONAL 2017; 2017:3706018. [PMID: 29138750 PMCID: PMC5613629 DOI: 10.1155/2017/3706018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Accepted: 08/09/2017] [Indexed: 12/18/2022]
Abstract
Aging is the principal risk factor for the development of Alzheimer's disease (AD). The hallmarks of AD are accumulation of the amyloid-β peptide 1-42 (Aβ42) and abnormal hyperphosphorylation of Tau (p-Tau) protein in different areas of the brain and, more recently reported, in the visual cortex. Recently, Aβ42 peptide overproduction has been involved in visual loss. Similar to AD, in normal aging, there is a significant amyloid deposition related to the overactivation of the aforementioned mechanisms. However, the mechanisms associated with visual loss secondary to age-induced visual cortex affectation are not completely understood. Young and aged mice were used as model to analyze the presence of Aβ42, p-Tau, glial-acidic fibrillary protein (GFAP), and presenilin-2, one of the main enzymes involved in Aβ42 production. Our results show a significant increase of Aβ42 deposition in aged mice in the following cells and/or tissues: endothelial cells and blood vessels and neurons of the visual cortex; they also show an increase of the expression of GFAP and presenilin-2 in this region. These results provide a comprehensive framework for the role of Aβ42 in visual loss due to inflammation present with aging and offer some clues for fruitful avenues for the study of healthy aging.
Collapse
Affiliation(s)
- Luis Fernando Hernández-Zimbrón
- Research Department, Asociación para Evitar la Ceguera en México, “Hospital Dr. Luis Sanchez Bulnes” IAP, 04030 México City, Mexico
| | - Montserrat Perez-Hernández
- Research Department, Asociación para Evitar la Ceguera en México, “Hospital Dr. Luis Sanchez Bulnes” IAP, 04030 México City, Mexico
- Divisón de Ciencias Biológicas de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Ciudad de México, Mexico
| | - Abigail Torres-Romero
- Research Department, Asociación para Evitar la Ceguera en México, “Hospital Dr. Luis Sanchez Bulnes” IAP, 04030 México City, Mexico
- Divisón de Ciencias Biológicas de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Ciudad de México, Mexico
| | - Elisa Gorostieta-Salas
- Neuroscience Division, Institute of Cellular Physiology, UNAM, Ciudad Universitaria, Ciudad de México, Mexico
| | - Roberto Gonzalez-Salinas
- Research Department, Asociación para Evitar la Ceguera en México, “Hospital Dr. Luis Sanchez Bulnes” IAP, 04030 México City, Mexico
| | - Rosario Gulias-Cañizo
- Research Department, Asociación para Evitar la Ceguera en México, “Hospital Dr. Luis Sanchez Bulnes” IAP, 04030 México City, Mexico
- Cell Biology Department, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, Mexico
| | - Hugo Quiroz-Mercado
- Research Department, Asociación para Evitar la Ceguera en México, “Hospital Dr. Luis Sanchez Bulnes” IAP, 04030 México City, Mexico
| | - Edgar Zenteno
- Department of Biochemistry, School of Medicine, UNAM, Ciudad Universitaria, México City, Mexico
| |
Collapse
|
21
|
Mehdinia A, Rostami S, Dadkhah S, Fumani NS. Simultaneous screening of homotaurine and taurine in marine macro-algae using liquid chromatography–fluorescence detection. JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2017. [DOI: 10.1007/s13738-017-1150-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
22
|
The effects of Chinese medicines on cAMP/PKA signaling in central nervous system dysfunction. Brain Res Bull 2017; 132:109-117. [PMID: 28438669 DOI: 10.1016/j.brainresbull.2017.04.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 04/11/2017] [Indexed: 01/06/2023]
Abstract
Neuropathological injury in the mammalian adult central nervous system (CNS) may cause axon disruption, neuronal death and lasting neurological deficits. Failure of axon regeneration is one of the major challenges for CNS functional recovery. Recently, the cAMP/PKA signaling pathway has been proven to be a critical regulator for neuronal regeneration, neuroplasticity, learning and memory. Also, previous studies have shown the effects of Chinese medicines on the prevention and treatment of CNS dysfunction mediated in part by cAMP/PKA signaling. In this review, the authors discuss current knowledge of the role of cAMP/PKA signaling pathway in neuronal regeneration and provide an overview of the Chinese medicines that may enable CNS functional recovery via this signaling pathway.
Collapse
|
23
|
Li F, Han G, Wu K. Tanshinone IIA Alleviates the AD Phenotypes in APP and PS1 Transgenic Mice. BIOMED RESEARCH INTERNATIONAL 2016; 2016:7631801. [PMID: 27274990 PMCID: PMC4870344 DOI: 10.1155/2016/7631801] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/02/2016] [Accepted: 03/15/2016] [Indexed: 12/31/2022]
Abstract
Therapeutic approach for Alzheimer's disease (AD) is still deficient. To find active compounds from herbal medicine is of interest in the alleviation of AD symptoms. This study aimed to investigate the protective effects of Tanshinone IIA (TIIA) on memory performance and synaptic plasticity in a transgenic AD model at the early phase. 25-100 mg/kg TIIA (intraperitoneal injection, i.p.) was administered to the six-month-old APP and PS1 transgenic mice for 30 consecutive days. After treatment, spatial memory, synaptic plasticity, and related mechanisms were investigated. Our result showed that memory impairment in AD mice was mitigated by 50 and 100 mg/kg TIIA treatments. Hippocampal long-term potentiation was impaired in AD model but rescued by 100 mg/kg TIIA treatment. Mechanically, TIIA treatment reduced the accumulations of beta-amyloid 1-42, C-terminal fragments (CTFs), and p-Tau in the AD model. TIIA did not affect basal BDNF but promoted depolarization-induced BDNF synthesis in the AD mice. Taken together, TIIA repairs hippocampal LTP and memory, likely, through facilitating the clearance of AD-related proteins and activating synaptic BDNF synthesis. TIIA might be a candidate drug for AD treatment.
Collapse
Affiliation(s)
- Fengling Li
- The Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, China
| | - Guosheng Han
- The Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, China
| | - Kexiang Wu
- The Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, China
| |
Collapse
|
24
|
Li F, Wu X, Li J, Niu Q. Ginsenoside Rg1 ameliorates hippocampal long-term potentiation and memory in an Alzheimer's disease model. Mol Med Rep 2016; 13:4904-10. [PMID: 27082952 DOI: 10.3892/mmr.2016.5103] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 03/02/2016] [Indexed: 11/06/2022] Open
Abstract
The complex etiopathogenesis of Alzheimer's disease (AD) has limited progression in the identification of effective therapeutic agents. Amyloid precursor protein (APP) and presenilin‑1 (PS1) are always overexpressed in AD, and are considered to be the initiators of the formation of β‑amyloid plaques and the symptoms of AD. In the present study, a transgenic AD model, constructed via the overexpression of APP and PS1, was used to verify the protective effects of ginsenoside Rg1 on memory performance and synaptic plasticity. AD mice (6‑month‑old) were treated via intraperitoneal injection of 0.1‑10 mg/kg ginsenoside Rg1. Long‑term memory, synaptic plasticity, and the levels of AD‑associated and synaptic plasticity‑associated proteins were measured following treatment. Memory was measured using a fear conditioning task and protein expression levels were investigated using western blotting. All the data was analyzed by one-way analysis of variance or t‑test. Following 30 days of consecutive treatment, memory in the AD mouse model was ameliorated in the 10 mg/kg ginsenoside Rg1 treatment group. As demonstrated by biochemical experiments, ginsenoside Rg1 treatment reduced the accumulations of β‑amyloid 1‑42 and phosphorylated (p)‑Tau in the AD model. Additionally, brain-derived neurotrophic factor (BDNF) and p‑TrkB synaptic plasticity‑associated proteins were upregulated following ginsenoside Rg1 application. Correspondingly, long‑term potentiation (LTP) was restored following ginsenoside Rg1 application in the AD mice model. Taken together, ginsenoside Rg1 repaired hippocampal LTP and memory, likely through facilitating the clearance of AD‑associated proteins and through activation of the BDNF‑TrkB pathway. Therefore, ginsenoside Rg1 may be a candidate drug for the treatment of AD.
Collapse
Affiliation(s)
- Fengling Li
- Department of Neurology, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, P.R. China
| | - Xiqing Wu
- Medical Imaging Center, Weifang Traditional Chinese Medicine Hospital, Weifang, Shandong 261041, P.R. China
| | - Jing Li
- Department of Orthopaedics Rehabilitation, Weifang Traditional Chinese Medicine Hospital, Weifang, Shandong 261041, P.R. China
| | - Qingliang Niu
- Medical Imaging Center, Weifang Traditional Chinese Medicine Hospital, Weifang, Shandong 261041, P.R. China
| |
Collapse
|
25
|
Matrine improves cognitive impairment and modulates the balance of Th17/Treg cytokines in a rat model of Aβ1-42-induced Alzheimer's disease. Cent Eur J Immunol 2016; 40:411-9. [PMID: 26862304 PMCID: PMC4737738 DOI: 10.5114/ceji.2015.56961] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 08/17/2015] [Indexed: 11/29/2022] Open
Abstract
Matrine (MAT) has been reported for its anti-inflammatory and neuroprotective effects. However, little is known about its effects on Th17/Treg cytokines and cognitive impairment in Alzheimer's disease (AD). In the present study, we injected Aβ1-42 to the hippocampus of the rat to induce AD. Three groups of the AD rats were treated with MAT (25, 100 or 200 mg/kg/day, respectively) by intraperitoneal injection for 5 weeks. Levels of Th17 cell cytokines [interleukin (IL)-17A and IL-23] and regulatory T (Treg) cell cytokines [transforming growth factor β (TGF-β) and IL-35] in homogenates of the brain cortex and hippocampus were measured using enzyme-linked immunosorbent assay (ELISA) kits. The mRNA expressions of Th17 cell specific transcription factor RORγt and Treg cell specific transcription factor Foxp3 in the brain cortex and hippocampus were quantified by real-time RT-PCR. Learning and memory ability of the rats were evaluated by Morris water maze test and novel object recognition test. ELISA detections showed the AD rats had increased levels of IL-17A and IL-23 as well as decreased levels of TGF-β and IL-35. Matrine (100 and 200 mg/kg/day) significantly reversed the alternations of Th17/Treg cytokines induced by Aβ1-42 injection, decreased RORγt mRNA expression, increased Foxp3 mRNA expression and improved the learning and memory ability in the AD rats. The findings demonstrated that the AD rats had imbalance of Th17/Treg cytokines in the brain. MAT could dose-dependently restore the balance of Th17/Treg cytokines and attenuate the cognitive impairment in AD rats.
Collapse
|
26
|
Abstract
Animal experiments and cross-sectional or prospective longitudinal research in human subjects suggest a role for nutrition in cognitive ageing. However, data from randomised controlled trials (RCT) that seek causal evidence for the impact of nutrients on cognitive ageing in humans often produce null results. Given that RCT test hypotheses in a rigorous fashion, one conclusion could be that the positive effects of nutrition on the aged brain observed in other study designs are spurious. On the other hand, it may be that the design of many clinical trials conducted thus far has been less than optimal. In the present review, we offer a blueprint for a more targeted approach to the design of RCT in nutrition, cognition and brain health in ageing that focuses on three key areas. First, the role of nutrition is more suited for the maintenance of health rather than the treatment of disease. Second, given that cognitive functions and brain regions vary in their susceptibility to ageing, those that especially deteriorate in senescence should be focal points in evaluating the efficacy of an intervention. Third, the outcome measures that assess change due to nutrition, especially in the cognitive domain, should not necessarily be the same neuropsychological tests used to assess gross brain damage or major pathological conditions. By addressing these three areas, we expect that clinical trials of nutrition, cognition and brain health in ageing will align more closely with other research in this field, and aid in revealing the true nature of nutrition's impact on the aged brain.
Collapse
|
27
|
Soheili M, Tavirani MR, Salami M. Lavandula angustifolia extract improves deteriorated synaptic plasticity in an animal model of Alzheimer's disease. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2015; 18:1147-52. [PMID: 26949505 PMCID: PMC4764119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
OBJECTIVES Neurodegenerative Alzheimer's disease (AD) is associated with profound deficits in synaptic transmission and synaptic plasticity. Long-term potentiation (LTP), an experimental form of synaptic plasticity, is intensively examined in hippocampus. In this study we evaluated the effect of aqueous extract of lavender (Lavandula angustifolia) on induction of LTP in the CA1 area of hippocampus. In response to stimulation of the Schaffer collaterals the baseline or tetanized field extracellular postsynaptic potentials (fEPSPs) were recorded in the CA1 area. MATERIALS AND METHODS The electrophysiological recordings were carried out in four groups of rats; two control groups including the vehicle (CON) and lavender (CE) treated rats and two Alzheimeric groups including the vehicle (ALZ) and lavender (AE) treated animals. RESULTS The extract inefficiently affected the baseline responses in the four testing groups. While the fEPSPs displayed a considerable LTP in the CON animals, no potentiation was evident in the tetanized responses in the ALZ rats. The herbal medicine effectively restored LTP in the AE group and further potentiated fEPSPs in the CE group. CONCLUSION The positive effect of the lavender extract on the plasticity of synaptic transmission supports its previously reported behavioral effects on improvement of impaired spatial memory in the Alzheimeric animals.
Collapse
Affiliation(s)
- Masoud Soheili
- International Branch, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Rezaei Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahmoud Salami
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran,Corresponding author: Mahmoud Salami. Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran. Tel: +98-913-3612920; Fax: +98-31-55621157;
| |
Collapse
|
28
|
Lee IS, Jung K, Kim IS, Lee H, Kim M, Yun S, Hwang K, Shin JE, Park KI. Human neural stem cells alleviate Alzheimer-like pathology in a mouse model. Mol Neurodegener 2015; 10:38. [PMID: 26293123 PMCID: PMC4546205 DOI: 10.1186/s13024-015-0035-6] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 08/10/2015] [Indexed: 12/14/2022] Open
Abstract
Background Alzheimer’s disease (AD) is an inexorable neurodegenerative disease that commonly occurs in the elderly. The cognitive impairment caused by AD is associated with abnormal accumulation of amyloid-β (Aβ) and hyperphosphorylated tau, which are accompanied by inflammation. Neural stem cells (NSCs) are self-renewing, multipotential cells that differentiate into distinct neural cells. When transplanted into a diseased brain, NSCs repair and replace injured tissues after migration toward and engraftment within lesions. We investigated the therapeutic effects in an AD mouse model of human NSCs (hNSCs) that derived from an aborted human fetal telencephalon at 13 weeks of gestation. Cells were transplanted into the cerebral lateral ventricles of neuron-specific enolase promoter-controlled APPsw-expressing (NSE/APPsw) transgenic mice at 13 months of age. Results Implanted cells extensively migrated and engrafted, and some differentiated into neuronal and glial cells, although most hNSCs remained immature. The hNSC transplantation improved spatial memory in these mice, which also showed decreased tau phosphorylation and Aβ42 levels and attenuated microgliosis and astrogliosis. The hNSC transplantation reduced tau phosphorylation via Trk-dependent Akt/GSK3β signaling, down-regulated Aβ production through an Akt/GSK3β signaling-mediated decrease in BACE1, and decreased expression of inflammatory mediators through deactivation of microglia that was mediated by cell-to-cell contact, secretion of anti-inflammatory factors generated from hNSCs, or both. The hNSC transplantation also facilitated synaptic plasticity and anti-apoptotic function via trophic supplies. Furthermore, the safety and feasibility of hNSC transplantation are supported. Conclusions These findings demonstrate the hNSC transplantation modulates diverse AD pathologies and rescue impaired memory via multiple mechanisms in an AD model. Thus, our data provide tangible preclinical evidence that human NSC transplantation could be a safe and versatile approach for treating AD patients. Electronic supplementary material The online version of this article (doi:10.1186/s13024-015-0035-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Il-Shin Lee
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 120-752, South Korea.
| | - Kwangsoo Jung
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 120-752, South Korea.
| | - Il-Sun Kim
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 120-752, South Korea.
| | - Haejin Lee
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 120-752, South Korea.
| | - Miri Kim
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 120-752, South Korea.
| | - Seokhwan Yun
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 120-752, South Korea.
| | - Kyujin Hwang
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 120-752, South Korea.
| | - Jeong Eun Shin
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 120-752, South Korea.
| | - Kook In Park
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 120-752, South Korea. .,Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 120-752, South Korea.
| |
Collapse
|
29
|
Bhattacharya S, Montag D. Acetylcholinesterase inhibitor modifications: a promising strategy to delay the progression of Alzheimer's disease. Neural Regen Res 2015; 10:43-5. [PMID: 25788916 PMCID: PMC4357112 DOI: 10.4103/1673-5374.150648] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2014] [Indexed: 11/21/2022] Open
Affiliation(s)
- Soumee Bhattacharya
- Leibniz Institute for Neurobiology, Neurogenetics Special Laboratory, Magdeburg, Germany
| | - Dirk Montag
- Leibniz Institute for Neurobiology, Neurogenetics Special Laboratory, Magdeburg, Germany
| |
Collapse
|
30
|
Shao H, Zhang Y, Dong Y, Yu B, Xia W, Xie Z. Chronic treatment with anesthetic propofol improves cognitive function and attenuates caspase activation in both aged and Alzheimer's disease transgenic mice. J Alzheimers Dis 2015; 41:499-513. [PMID: 24643139 DOI: 10.3233/jad-132792] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
There is a need to seek new treatment(s) for Alzheimer's disease (AD). A recent study showed that AD patients may have decreased levels of functional GABA receptors. Propofol, a commonly used anesthetic, is a GABA receptor agonist. We therefore set out to perform a proof of concept study to determine whether chronic treatment with propofol (50 mg/kg/week) can improve cognitive function in both aged wild-type (WT) and AD transgenic (Tg) mice. Propofol was administrated to the WT and AD Tg mice once a week for 8 or 12 weeks, respectively. Morris water maze was used to assess the cognitive function of the mice following the propofol treatment. Activation of caspase-3, caspase-9, and caspase-8 was investigated using western blot analysis at the end of the propofol treatment. In the mechanistic studies, effects of propofol, amyloid-β protein (Aβ), and GABA receptor antagonist flumazenil on caspase-3 activation and opening of the mitochondrial permeability transition pore were assessed in H4 human neuroglioma and mouse neuroblastoma cells by western blot analysis and flow cytometry. Here we showed that the propofol treatment improved cognitive function and attenuated brain caspase-3 and caspase-9 activation in both aged WT and AD Tg mice. Propofol attenuated Aβ-induced caspase-3 activation and opening of the mitochondrial permeability transition pore in the cells, and flumazenil inhibited the propofol's effects. These results suggested that propofol might improve cognitive function via attenuating the Aβ-induced mitochondria dysfunction and caspase activation, which explored the potential that anesthetic propofol could improve cognitive function in elderly and AD patients.
Collapse
Affiliation(s)
- Haijun Shao
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.,Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Yiying Zhang
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Yuanlin Dong
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Buwei Yu
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Weiming Xia
- Department of Veterans Affairs, Medical Research and Development Service and Geriatric Research, Education and Clinical Center, Bedford, MA, USA
| | - Zhongcong Xie
- Geriatric Anesthesia Research Unit, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
31
|
Kim HY, Kim HV, Yoon JH, Kang BR, Cho SM, Lee S, Kim JY, Kim JW, Cho Y, Woo J, Kim Y. Taurine in drinking water recovers learning and memory in the adult APP/PS1 mouse model of Alzheimer's disease. Sci Rep 2014; 4:7467. [PMID: 25502280 PMCID: PMC4264000 DOI: 10.1038/srep07467] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 11/25/2014] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a lethal progressive neurological disorder affecting the memory. Recently, US Food and Drug Administration mitigated the standard for drug approval, allowing symptomatic drugs that only improve cognitive deficits to be allowed to accelerate on to clinical trials. Our study focuses on taurine, an endogenous amino acid found in high concentrations in humans. It has demonstrated neuroprotective properties against many forms of dementia. In this study, we assessed cognitively enhancing property of taurine in transgenic mouse model of AD. We orally administered taurine via drinking water to adult APP/PS1 transgenic mouse model for 6 weeks. Taurine treatment rescued cognitive deficits in APP/PS1 mice up to the age-matching wild-type mice in Y-maze and passive avoidance tests without modifying the behaviours of cognitively normal mice. In the cortex of APP/PS1 mice, taurine slightly decreased insoluble fraction of Aβ. While the exact mechanism of taurine in AD has not yet been ascertained, our results suggest that taurine can aid cognitive impairment and may inhibit Aβ-related damages.
Collapse
Affiliation(s)
- Hye Yun Kim
- 1] Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea [2] Biological Chemistry Program, Korea University of Science and Technology, Daejeon, Republic of Korea [3] Department of Biochemistry and Biomedical Sciences, Seoul National University, College of Medicine, Seoul, Republic of Korea
| | - Hyunjin V Kim
- 1] Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea [2] Biological Chemistry Program, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Jin H Yoon
- 1] Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea [2] Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, U.S.A
| | - Bo Ram Kang
- 1] Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea [2] Biological Chemistry Program, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Soo Min Cho
- 1] Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea [2] Biological Chemistry Program, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Sejin Lee
- 1] Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea [2] Biological Chemistry Program, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Ji Yoon Kim
- 1] Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea [2] Biological Chemistry Program, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Joo Won Kim
- 1] Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea [2] Department of Applied Chemistry, Dongduk Women's University, Seoul, Republic of Korea
| | - Yakdol Cho
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Jiwan Woo
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - YoungSoo Kim
- 1] Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea [2] Biological Chemistry Program, Korea University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
32
|
Physical exercise neuroprotects ovariectomized 3xTg-AD mice through BDNF mechanisms. Psychoneuroendocrinology 2014; 45:154-66. [PMID: 24845186 DOI: 10.1016/j.psyneuen.2014.03.021] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 03/29/2014] [Accepted: 03/30/2014] [Indexed: 11/22/2022]
Abstract
Postmenopausal women may be more vulnerable to cognitive loss and Alzheimer's disease (AD) than premenopausal women because of their deficiency in estrogens, in addition to their usually older age. Aerobic physical exercise has been proposed as a therapeutic approach for maintaining health and well-being in postmenopausal women, and for improving brain health and plasticity in populations at high risk for AD. To study the neuroprotective mechanisms of physical exercise in a postmenopausal animal model, we submitted previously ovariectomized, six-month old non-transgenic and 3xTg-AD mice to three months of voluntary exercise in a running wheel. At nine months of age, we observed lower grip strength and some exacerbation of the behavioral and psychological symptoms of dementia (BPSD)-like involving active exploratory activities. A similar major cognitive impairment was observed of ovariectomized 3xTg-AD mice in comparison with sham-operated 3xTg-AD mice. A reduction of bodily fitness and lack of retention of memory were observed in the ovariectomized non-transgenic mice. Physical exercise protected against all deleterious behaviors and normalized learning and memory. It also protected against body frailty, as expected. Analyses of hippocampal key markers of antioxidant and neuroplasticity signaling pathways, showed that ovariectomy impairs the activation of CREB through physical exercise. Furthermore, molecular and behavioral correlates suggested a central role of BDNF in the neuroprotection mediated by physical exercise therapy against apathy and memory loss induced by ovariectomy and the AD-genotype.
Collapse
|
33
|
SCHUCK PATRÍCIAFERNANDA. Mitochondrial DNA, anti-tuberculosis drugs-induced hepatotoxicity and Alzheimer's disease. AN ACAD BRAS CIENC 2014. [DOI: 10.1590/0001-37652014862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
34
|
Hajimehdipoor H, Mosaddegh M, Naghibi F, Haeri A, Hamzeloo-Moghadam M. Natural sesquiterpen lactones as acetylcholinesterase inhibitors. AN ACAD BRAS CIENC 2014; 86:801-806. [PMID: 24838542 DOI: 10.1590/0001-3765201420130005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 10/14/2013] [Indexed: 01/30/2023] Open
Abstract
Background and the purpose of the study: The amount of elder people who suffer from Alzheimer disease is continuously increasing every year. Cholinesterase inhibitors have shown to be effective in alleviating the symptoms of the disease, thus opening a field of research for these treatments. Herbal products, owning a reputation as effective agents in many biological studies are now drawing attention for inhibiting acetylcholinesterase, in other words, Alzheimer disease. In the present study, the ability of three sesquiterpene lactones from Inula oculus-christi and I. aucheriana to inhibit AChE has been evaluated through Ellman assay. Materials and Methods: Gaillardin and pulchellin C were obtained from I. oculus-christi and britannin from I. aucheriana by chromatographic methods. They were dissolved in methanol in concentration of 3 mg/mL and the AChEI activity of the compounds was determined by Ellman method using Acethylthiocholine iodide as the substrate and 5, 5'-dithiobis-2-nitrobenzoic acid as the reagent, in 96-well plates at 405 nm. Results: AChEI activity of the examined compounds was obtained as 67.0, 25.2 and 10.9% in concentration of 300 µg/L for gaillardin, britannin and pulchellin C, respectively. Conclusion: Among the three sesquiterpene lactones, gaillardin with 67% inhibition of AChE could be considered a good candidate for future Alzheimer studies.
Collapse
Affiliation(s)
- Homa Hajimehdipoor
- Department of Traditional Pharmacy, School of Traditional Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahmoud Mosaddegh
- Department of Traditional Pharmacy, School of Traditional Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzaneh Naghibi
- Department of Traditional Pharmacy, School of Traditional Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Haeri
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Hamzeloo-Moghadam
- Traditional Medicine and Materia Medica Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
Davis PR, Head E. Prevention approaches in a preclinical canine model of Alzheimer's disease: benefits and challenges. Front Pharmacol 2014; 5:47. [PMID: 24711794 PMCID: PMC3968758 DOI: 10.3389/fphar.2014.00047] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 02/28/2014] [Indexed: 12/30/2022] Open
Abstract
Aged dogs spontaneously develop many features of human aging and Alzheimer's disease (AD) including cognitive decline and neuropathology. In this review, we discuss age-dependent learning tasks, memory tasks, and functional measures that can be used in aged dogs for sensitive treatment outcome measures. Neuropathology that is linked to cognitive decline is described along with examples of treatment studies that show reduced neuropathology in aging dogs (dietary manipulations, behavioral enrichment, immunotherapy, and statins). Studies in canine show that multi-targeted approaches may be more beneficial than single pathway manipulations (e.g., antioxidants combined with behavioral enrichment). Aging canine studies show good predictive validity for human clinical trials outcomes (e.g., immunotherapy) and several interventions tested in dogs strongly support a prevention approach (e.g., immunotherapy and statins). Further, dogs are ideally suited for prevention studies as they the age because onset of cognitive decline and neuropathology strongly support longitudinal interventions that can be completed within a 3-5 year period. Disadvantages to using the canine model are that they lengthy, use labor-intensive comprehensive cognitive testing, and involve costly housing (almost as high as that of non-human primates). However, overall, using the dog as a preclinical model for testing preventive approaches for AD may complement work in rodents and non-human primates.
Collapse
Affiliation(s)
- Paulina R Davis
- Sanders-Brown Center on Aging, University of Kentucky Lexington, KY, USA ; Department of Molecular and Biomedical Pharmacology, University of Kentucky Lexington, KY, USA
| | - Elizabeth Head
- Sanders-Brown Center on Aging, University of Kentucky Lexington, KY, USA ; Department of Molecular and Biomedical Pharmacology, University of Kentucky Lexington, KY, USA
| |
Collapse
|
36
|
Wyse RD, Dunbar GL, Rossignol J. Use of genetically modified mesenchymal stem cells to treat neurodegenerative diseases. Int J Mol Sci 2014; 15:1719-45. [PMID: 24463293 PMCID: PMC3958818 DOI: 10.3390/ijms15021719] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 12/18/2013] [Accepted: 01/14/2014] [Indexed: 01/01/2023] Open
Abstract
The transplantation of mesenchymal stem cells (MSCs) for treating neurodegenerative disorders has received growing attention recently because these cells are readily available, easily expanded in culture, and when transplanted, survive for relatively long periods of time. Given that such transplants have been shown to be safe in a variety of applications, in addition to recent findings that MSCs have useful immunomodulatory and chemotactic properties, the use of these cells as vehicles for delivering or producing beneficial proteins for therapeutic purposes has been the focus of several labs. In our lab, the use of genetic modified MSCs to release neurotrophic factors for the treatment of neurodegenerative diseases is of particular interest. Specifically, glial cell-derived neurotrophic factor (GDNF), nerve growth factor (NGF), and brain derived neurotrophic factor (BDNF) have been recognized as therapeutic trophic factors for Parkinson's, Alzheimer's and Huntington's diseases, respectively. The aim of this literature review is to provide insights into: (1) the inherent properties of MSCs as a platform for neurotrophic factor delivery; (2) the molecular tools available for genetic manipulation of MSCs; (3) the rationale for utilizing various neurotrophic factors for particular neurodegenerative diseases; and (4) the clinical challenges of utilizing genetically modified MSCs.
Collapse
Affiliation(s)
- Robert D Wyse
- Field Neurosciences Institute Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, Central Michigan University, Mount Pleasant, MI 48859, USA.
| | - Gary L Dunbar
- Field Neurosciences Institute Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, Central Michigan University, Mount Pleasant, MI 48859, USA.
| | - Julien Rossignol
- Field Neurosciences Institute Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, Central Michigan University, Mount Pleasant, MI 48859, USA.
| |
Collapse
|
37
|
Chen JF. Adenosine receptor control of cognition in normal and disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2014; 119:257-307. [PMID: 25175970 DOI: 10.1016/b978-0-12-801022-8.00012-x] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Adenosine and adenosine receptors (ARs) are increasingly recognized as important therapeutic targets for controlling cognition under normal and disease conditions for its dual roles of neuromodulation as well as of homeostatic function in the brain. This chapter first presents the unique ability of adenosine, by acting on the inhibitory A1 and facilitating A2A receptor, to integrate dopamine, glutamate, and BNDF signaling and to modulate synaptic plasticity (e.g., long-term potentiation and long-term depression) in brain regions relevant to learning and memory, providing the molecular and cellular bases for adenosine receptor (AR) control of cognition. This led to the demonstration of AR modulation of social recognition memory, working memory, reference memory, reversal learning, goal-directed behavior/habit formation, Pavlovian fear conditioning, and effort-related behavior. Furthermore, human and animal studies support that AR activity can also, through cognitive enhancement and neuroprotection, reverse cognitive impairments in animal models of Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease, and schizophrenia. Lastly, epidemiological evidence indicates that regular human consumption of caffeine, the most widely used psychoactive drug and nonselective AR antagonists, is associated with the reduced cognitive decline in aging and AD patients, and with the reduced risk in developing PD. Thus, there is a convergence of the molecular studies revealing AR as molecular targets for integrating neurotransmitter signaling and controlling synaptic plasticity, with animal studies demonstrating the strong procognitive impact upon AR antagonism in normal and disease brains and with epidemiological and clinical evidences in support of caffeine and AR drugs for therapeutic modulation of cognition. Since some of adenosine A2A receptor antagonists are already in phase III clinical trials for motor benefits in PD patients with remarkable safety profiles, additional animal and human studies to better understand the mechanism underlying the AR-mediated control of cognition under normal and disease conditions will provide the required rationale to stimulate the necessary clinical investigation to rapidly translate adenosine and AR drug as a novel strategy to control memory impairment in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jiang-Fan Chen
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA; The Molecular Medicine Institute, Wenzhou Medical University, Wenzhou, Zhejiang, PR China.
| |
Collapse
|
38
|
Burk JA. Roles of cholinergic receptors during attentional modulation of cue detection. World J Pharmacol 2013; 2:84-91. [DOI: 10.5497/wjp.v2.i4.84] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Revised: 08/17/2013] [Accepted: 09/13/2013] [Indexed: 02/06/2023] Open
Abstract
Basal forebrain corticopetal cholinergic neurons are known to be necessary for normal attentional processing. Alterations of cholinergic system functioning have been associated with several neuropsychiatric diseases, such as Alzheimer’s disease and schizophrenia, in which attentional dysfunction is thought to be a key contributing factor. Loss of cortical cholinergic inputs impairs performance in attention-demanding tasks. Moreover, measures of acetylcholine with microdialysis and, more recently, of choline with enzyme-coated microelectrodes have begun to elucidate the precise cognitive demands that activate the cholinergic system on distinct time scales. However, the receptor actions following acetylcholine release under attentionally-challenging conditions are only beginning to be understood. The present review is designed to summarize the evidence regarding the actions of acetylcholine at muscarinic and nicotinic receptors under cognitively challenging conditions in order to evaluate the functions mediated by these two different cholinergic receptor classes. Moreover, evidence that supports beneficial effects of muscarinic muscarinic-1 receptor agonists and selective nicotinic receptor subtype agonists for cognitive processing will be discussed. Finally, some challenges and limitations of targeting the cholinergic system for treating cognitive deficits along with future research directions will be mentioned. In conclusion, multiple aspects of cholinergic neurotransmission must be considered when attempting to restore function of this neuromodulatory system.
Collapse
|
39
|
Ommaya AK, Adams KM, Allman RM, Collins EG, Cooper RA, Dixon CE, Fishman PS, Henry JA, Kardon R, Kerns RD, Kupersmith J, Lo A, Macko R, McArdle R, McGlinchey RE, McNeil MR, O'Toole TP, Peckham PH, Tuszynski MH, Waxman SG, Wittenberg GF. Guest editorial: Opportunities in rehabilitation research. ACTA ACUST UNITED AC 2013; 50:vii-xxxii. [PMID: 24203548 DOI: 10.1682/jrrd.2012.09.0167] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
40
|
Abstract
Alzheimer disease represents an insidious impairment of intellect and emotional well-being. However, recent advances in biochemical pathology and human genetics offer promise that effective therapeutic agents may soon be developed.
Collapse
Affiliation(s)
- Dennis Selkoe
- Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts 02115, USA.
| | | | | |
Collapse
|
41
|
Enciu AM, Popescu BO. Is there a causal link between inflammation and dementia? BIOMED RESEARCH INTERNATIONAL 2013; 2013:316495. [PMID: 23841061 PMCID: PMC3690213 DOI: 10.1155/2013/316495] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Accepted: 05/20/2013] [Indexed: 12/22/2022]
Abstract
Neuroinflammation is a constant event in Alzheimer's disease (AD), but the current knowledge is insufficient to state whether inflammation is a cause, a promoter, or simply a secondary phenomenon in this inexorably progressive ailment. In the current paper, we review research data showing that inflammation is not a prerequisite for onset of dementia, and, although it may worsen the course of the disease, recent evidence shows that chronic inhibition of inflammatory pathways is not necessarily beneficial for patients. Prospective clinical trials with anti-inflammatory drugs failed to stop disease progression, measurements of inflammatory markers in serum and cerebrospinal fluid of patients yielded contradictory results, and recent bench research proved undoubtedly that neuroinflammation has a protective side as well. Knockout animal models for TNFRs or ILRs do not seem to prevent the pathology or the cognitive decline, but quite the contrary. In AD, the therapeutic intervention on inflammatory pathways still has a research future, but its targets probably need reevaluation.
Collapse
Affiliation(s)
- Ana-Maria Enciu
- Department of Cellular and Molecular Medicine, School of Medicine, “Carol Davila” University of Medicine and Pharmacy, 8 Eroilor Sanitari, District 5, Bucharest 050474, Romania
| | - Bogdan O. Popescu
- Department of Neurology, Colentina Clinical Hospital (CDPC), School of Medicine, “Carol Davila” University of Medicine and Pharmacy, 19-21 Soseaua Stefan cel Mare, District 2, Bucharest 020125, Romania
- Laboratory of Molecular Medicine, “Victor Babeş” National Institute of Pathology, 99-101 Splaiul Independenţei, District 5, Bucharest 050096, Romania
| |
Collapse
|
42
|
Abstract
Alzheimer's disease (AD) is an urgent public health challenge that is rapidly approaching epidemic proportions. New therapies that defer or prevent the onset, delay the decline, or improve the symptoms are urgently needed. All phase 3 drug development programs for disease-modifying agents have failed thus far. New approaches to drug development are needed. Translational neuroscience focuses on the linkages between basic neuroscience and the development of new diagnostic and therapeutic products that will improve the lives of patients or prevent the occurrence of brain disorders. Translational neuroscience includes new preclinical models that may better predict human efficacy and safety, improved clinical trial designs and outcomes that will accelerate drug development, and the use of biomarkers to more rapidly provide information regarding the effects of drugs on the underlying disease biology. Early translational research is complemented by later stage translational approaches regarding how best to use evidence to impact clinical practice and to assess the influence of new treatments on the public health. Funding of translational research is evolving with an increased emphasis on academic and NIH involvement in drug development. Translational neuroscience provides a framework for advancing development of new therapies for AD patients.
Collapse
|
43
|
Lin LF, Liao MJ, Xue XY, Zhang W, Yan L, Cai L, Zhou XW, Zhou X, Luo HM. Combination of Aβ clearance and neurotrophic factors as a potential treatment for Alzheimer's disease. Neurosci Bull 2013; 29:111-20. [PMID: 23179066 PMCID: PMC5561854 DOI: 10.1007/s12264-012-1287-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 06/12/2012] [Indexed: 12/31/2022] Open
Abstract
There is no effective drug to treat Alzheimer's disease (AD), a neurodegenerative disease affecting an estimated 30 million people around the world. Strongly supported by preclinical and clinical studies, amyloid-beta (Aβ) may be a target for developing drugs against AD. Meanwhile, the fact that localized neuronal death/loss and synaptic impairment occur in AD should also be considered. Neuronal regeneration, which does not occur normally in the mammalian central nervous system, can be promoted by neurotrophic factors (NTFs). Evidence from clinical trials has shown that both Aβ clearance and NTFs are potentially effective in treating AD, thus a new approach combining Aβ clearance and administration of NTFs may be an effective therapeutic strategy.
Collapse
Affiliation(s)
- Lian-Feng Lin
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
| | - Min-Jing Liao
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
- Department of Laboratory Medicine, Medical College of Hunan Normal University, Changsha, 410013 China
| | - Xiao-Yan Xue
- Ganzhou People’s Hospital, Ganzhou, 341000 China
| | - Wei Zhang
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
| | - Li Yan
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
| | - Liang Cai
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
| | - Xiao-Wen Zhou
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
| | - Xing Zhou
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
| | - Huan-Min Luo
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
- Institute of Brain Sciences, School of Medicine, Jinan University, Guangzhou, 510632 China
- The Joint Laboratory of Brain Function and Health, Jinan University and The University of Hong Kong, Jinan University, Guangzhou, 510632 China
| |
Collapse
|
44
|
Mullane K, Williams M. Alzheimer's therapeutics: continued clinical failures question the validity of the amyloid hypothesis-but what lies beyond? Biochem Pharmacol 2012. [PMID: 23178653 DOI: 10.1016/j.bcp.2012.11.014] [Citation(s) in RCA: 147] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The worldwide incidence of Alzheimer's disease (AD) is increasing with estimates that 115 million individuals will have AD by 2050, creating an unsustainable healthcare challenge due to a lack of effective treatment options highlighted by multiple clinical failures of agents designed to reduce the brain amyloid burden considered synonymous with the disease. The amyloid hypothesis that has been the overarching focus of AD research efforts for more than two decades has been questioned in terms of its causality but has not been unequivocally disproven despite multiple clinical failures, This is due to issues related to the quality of compounds advanced to late stage clinical trials and the lack of validated biomarkers that allow the recruitment of AD patients into trials before they are at a sufficiently advanced stage in the disease where therapeutic intervention is deemed futile. Pursuit of a linear, reductionistic amyloidocentric approach to AD research, which some have compared to a religious faith, has resulted in other, equally plausible but as yet unvalidated AD hypotheses being underfunded leading to a disastrous roadblock in the search for urgently needed AD therapeutics. Genetic evidence supporting amyloid causality in AD is reviewed in the context of the clinical failures, and progress in tau-based and alternative approaches to AD, where an evolving modus operandi in biomedical research fosters excessive optimism and a preoccupation with unproven, and often ephemeral, biomarker/genome-based approaches that override transparency, objectivity and data-driven decision making, resulting in low probability environments where data are subordinate to self propagating hypotheses.
Collapse
|
45
|
Abstract
Remarkable advances in unraveling the biological underpinnings of Alzheimer disease (AD) have occurred during the last 25 years. Despite this, we have made only the smallest of dents in the development of truly disease-modifying treatments. What will change over the next 10 years? While the answer is not clear, we make several predictions on the state of the field in 2020, based on the rich knowledge described in the other contributions in this collection. As such, our predictions represent some of the principal unresolved questions that we believe deserve special investigative attention in the coming decade.
Collapse
Affiliation(s)
- David M Holtzman
- Department of Neurology, Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, Missouri 63110, USA.
| | | | | |
Collapse
|
46
|
Vellas B, Coley N, Ousset PJ, Berrut G, Dartigues JF, Dubois B, Grandjean H, Pasquier F, Piette F, Robert P, Touchon J, Garnier P, Mathiex-Fortunet H, Andrieu S. Long-term use of standardised ginkgo biloba extract for the prevention of Alzheimer's disease (GuidAge): a randomised placebo-controlled trial. Lancet Neurol 2012; 11:851-9. [DOI: 10.1016/s1474-4422(12)70206-5] [Citation(s) in RCA: 274] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
47
|
Mayeux R, Stern Y. Epidemiology of Alzheimer disease. Cold Spring Harb Perspect Med 2012; 2:cshperspect.a006239. [PMID: 22908189 DOI: 10.1101/cshperspect.a006239] [Citation(s) in RCA: 589] [Impact Index Per Article: 49.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The global prevalence of dementia has been estimated to be as high as 24 million, and is predicted to double every 20 years until at least 2040. As the population worldwide continues to age, the number of individuals at risk will also increase, particularly among the very old. Alzheimer disease is the leading cause of dementia beginning with impaired memory. The neuropathological hallmarks of Alzheimer disease include diffuse and neuritic extracellular amyloid plaques in brain that are frequently surrounded by dystrophic neurites and intraneuronal neurofibrillary tangles. The etiology of Alzheimer disease remains unclear, but it is likely to be the result of both genetic and environmental factors. In this review we discuss the prevalence and incidence rates, the established environmental risk factors, and the protective factors, and briefly review genetic variants predisposing to disease.
Collapse
Affiliation(s)
- Richard Mayeux
- Gertrude H. Sergievsky Center, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY 10032, USA
| | | |
Collapse
|