1
|
Xu Y, Filice CT, Leonenko Z. Protective effect of trehalose sugar on amyloid-membrane interactions using BLM electrophysiology. Biophys J 2024; 123:1690-1704. [PMID: 38751113 PMCID: PMC11213996 DOI: 10.1016/j.bpj.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/10/2024] [Accepted: 05/13/2024] [Indexed: 05/30/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by dementia and memory loss in the elderly population. The amyloid-β peptide (Aβ) is one of the main pathogenic factors in AD and is known to cause damage to neuronal cellular membranes. There is no cure currently available for AD, and new approaches, including preventive strategies, are highly desirable. In this work, we explore the possibility of protecting neuronal membranes from amyloid-induced damage with naturally existing sugar trehalose. Trehalose has been shown to protect plant cellular membranes in extreme conditions and modify Aβ misfolding. We hypothesize that trehalose can protect the neuronal membrane from amyloid toxicity. In this work, we studied the protective effect of trehalose against Aβ1-42-induced damage in model lipid membranes (DPPC/POPC/cholesterol) using atomic force microscopy and black lipid membrane electrophysiology. Our results demonstrate that Aβ1-42 damaged membranes and led to ionic current leakage across these membranes due to the formation of various defects and pores. The presence of trehalose reduced the ion current across membranes caused by Aβ1-42 peptide damage, thus efficiently protecting the membranes. These findings suggest that the trehalose sugar can potentially be useful in protecting neuronal membranes against amyloid toxicity in AD.
Collapse
Affiliation(s)
- Yue Xu
- Department of Physics & Astronomy, University of Waterloo, Waterloo, ON, Canada
| | - Carina Teresa Filice
- Department of Biology, University of Waterloo, Waterloo, ON, Canada; Waterloo Institute for Nanotechnology, Waterloo, ON, Canada
| | - Zoya Leonenko
- Department of Physics & Astronomy, University of Waterloo, Waterloo, ON, Canada; Department of Biology, University of Waterloo, Waterloo, ON, Canada; Waterloo Institute for Nanotechnology, Waterloo, ON, Canada.
| |
Collapse
|
2
|
Sukreet S, Rafii MS, Rissman RA. From understanding to action: Exploring molecular connections of Down syndrome to Alzheimer's disease for targeted therapeutic approach. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12580. [PMID: 38623383 PMCID: PMC11016820 DOI: 10.1002/dad2.12580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 04/17/2024]
Abstract
Down syndrome (DS) is caused by a third copy of chromosome 21. Alzheimer's disease (AD) is a neurodegenerative condition characterized by the deposition of amyloid-beta (Aβ) plaques and neurofibrillary tangles in the brain. Both disorders have elevated Aβ, tau, dysregulated immune response, and inflammation. In people with DS, Hsa21 genes like APP and DYRK1A are overexpressed, causing an accumulation of amyloid and neurofibrillary tangles, and potentially contributing to an increased risk of AD. As a result, people with DS are a key demographic for research into AD therapeutics and prevention. The molecular links between DS and AD shed insights into the underlying causes of both diseases and highlight potential therapeutic targets. Also, using biomarkers for early diagnosis and treatment monitoring is an active area of research, and genetic screening for high-risk individuals may enable earlier intervention. Finally, the fundamental mechanistic parallels between DS and AD emphasize the necessity for continued research into effective treatments and prevention measures for DS patients at risk for AD. Genetic screening with customized therapy approaches may help the DS population in current clinical studies and future biomarkers.
Collapse
Affiliation(s)
- Sonal Sukreet
- Department of NeurosciencesUniversity of California‐San DiegoLa JollaCaliforniaUSA
| | - Michael S. Rafii
- Department of Neurology, Alzheimer's Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Robert A. Rissman
- Department of NeurosciencesUniversity of California‐San DiegoLa JollaCaliforniaUSA
- Department Physiology and Neuroscience, Alzheimer’s Therapeutic Research InstituteKeck School of Medicine of the University of Southern CaliforniaSan DiegoCaliforniaUSA
| |
Collapse
|
3
|
Niu B, Pan T, Xiao Y, Wang H, Zhu J, Tian F, Lu W, Chen W. The therapeutic potential of dietary intervention: based on the mechanism of a tryptophan derivative-indole propionic acid on metabolic disorders. Crit Rev Food Sci Nutr 2024:1-20. [PMID: 38189263 DOI: 10.1080/10408398.2023.2299744] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Tryptophan (TRP) contributes to individual immune homeostasis and good condition via three complex metabolism pathways (5-hydroxytryptamine (5-HT), kynurenine (KP), and gut microbiota pathway). Indole propionic acid (IPA), one of the TRP derivatives of the microbiota pathway, has raised more attention because of its impact on metabolic disorders. Here, we retrospect increasing evidence that TRP metabolites/IPA derived from its proteolysis impact host health and disease. IPA can activate the immune system through aryl hydrocarbon receptor (AHR) and/or Pregnane X receptor (PXR) as a vital mediator among diet-caused host and microbe cross-talk. Different levels of IPA in systemic circulation can predict the risk of NAFLD, T2DM, and CVD. IPA is suggested to alleviate cognitive impairment from oxidative damage, reduce gut inflammation, inhibit lipid accumulation and attenuate the symptoms of NAFLD, putatively enhance the intestinal epithelial barrier, and maintain intestinal homeostasis. Now, we provide a general description of the relationships between IPA and various physiological and pathological processes, which support an opportunity for diet intervention for metabolic diseases.
Collapse
Affiliation(s)
- Ben Niu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Tong Pan
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yue Xiao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hongchao Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jinlin Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| |
Collapse
|
4
|
Extensive Summary of the Important Roles of Indole Propionic Acid, a Gut Microbial Metabolite in Host Health and Disease. Nutrients 2022; 15:nu15010151. [PMID: 36615808 PMCID: PMC9824871 DOI: 10.3390/nu15010151] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 12/30/2022] Open
Abstract
Increasing evidence suggests that metabolites produced by the gut microbiota play a crucial role in host-microbe interactions. Dietary tryptophan ingested by the host enters the gut, where indole-like metabolites such as indole propionic acid (IPA) are produced under deamination by commensal bacteria. Here, we summarize the IPA-producing bacteria, dietary patterns on IPA content, and functional roles of IPA in various diseases. IPA can not only stimulate the expression of tight junction (TJ) proteins to enhance gut barrier function and inhibit the penetration of toxic factors, but also modulate the immune system to exert anti-inflammatory and antioxidant effects to synergistically regulate body physiology. Moreover, IPA can act on target organs through blood circulation to form the gut-organ axis, which helps maintain systemic homeostasis. IPA shows great potential for the diagnosis and treatment of various clinical diseases, such as NAFLD, Alzheimer's disease, and breast cancer. However, the therapeutic effect of IPA depends on dose, target organ, or time. In future studies, further work should be performed to explore the effects and mechanisms of IPA on host health and disease to further improve the existing treatment program.
Collapse
|
5
|
Natsume S, Baba H, Maeshima H, Saida T, Yoshinari N, Shimizu K, Suzuki T. Clinical course and serum amyloid β levels in elderly patients with major depressive disorder. J Affect Disord 2022; 315:156-161. [PMID: 35932935 DOI: 10.1016/j.jad.2022.07.073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/26/2022] [Accepted: 07/30/2022] [Indexed: 10/16/2022]
Abstract
BACKGROUND Depression is known to be a risk factor for Alzheimer's disease (AD). Changes in amyloid β protein (Aβ) metabolism have been speculated as a factor contributing to the transition from depression to AD. The aim of this study is to reveal the time course and state-dependency of Aβ metabolism. METHODS Serum Aβ levels in 277 elderly (≥60 years) patients with depression (both early- and late-onset) were measured at admission, immediately after remission, and 1 year after remission, and compared them with 178 healthy subjects. RESULTS The analysis revealed decreased Aβ42 levels and increased Aβ42/40 ratios in elderly patients with depression at admission compared with healthy subjects. These changes in the acute phase of depression were not normalized immediately after remission; however, they recovered to healthy levels 1 year after remission. LIMITATIONS There is a possibility that the results may be influenced by antidepressants. CONCLUSIONS These results suggest that altered Aβ metabolism caused by depression may ameliorate, although after a lengthy period of time after remission. Our findings also suggest that the AD-related pathological changes caused or increased by depression can be reduced by maintaining remission for an extended period of time.
Collapse
Affiliation(s)
- Shuntaro Natsume
- Department of Psychiatry & Behavioral Science, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hajime Baba
- Department of Psychiatry & Behavioral Science, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Psychiatry, Juntendo Koshigaya Hospital, Juntendo University Faculty of Medicine, Saitama, Japan.
| | - Hitoshi Maeshima
- Department of Psychiatry & Behavioral Science, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Psychiatry, Juntendo Koshigaya Hospital, Juntendo University Faculty of Medicine, Saitama, Japan
| | - Takao Saida
- Department of Psychiatry & Behavioral Science, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Naoto Yoshinari
- Department of Psychiatry & Behavioral Science, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Psychiatry, Juntendo Koshigaya Hospital, Juntendo University Faculty of Medicine, Saitama, Japan
| | - Kentaro Shimizu
- Department of Psychiatry & Behavioral Science, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Toshihito Suzuki
- Department of Psychiatry & Behavioral Science, Juntendo University Graduate School of Medicine, Tokyo, Japan; Department of Psychiatry, Juntendo Koshigaya Hospital, Juntendo University Faculty of Medicine, Saitama, Japan
| |
Collapse
|
6
|
Liu Y, Zhang S, He B, Chen L, Ke D, Zhao S, Zhang Y, Wei W, Xu Z, Xu Z, Yin Y, Mo W, Li Y, Gao Y, Li S, Wang W, Yu H, Wu D, Pi G, Jiang T, Deng M, Xiong R, Lei H, Tian N, He T, Sun F, Zhou Q, Wang X, Ye J, Li M, Hu N, Song G, Peng W, Zheng C, Zhang H, Wang JZ. Periphery Biomarkers for Objective Diagnosis of Cognitive Decline in Type 2 Diabetes Patients. Front Cell Dev Biol 2021; 9:752753. [PMID: 34746146 PMCID: PMC8564071 DOI: 10.3389/fcell.2021.752753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/23/2021] [Indexed: 12/03/2022] Open
Abstract
Introduction: Type 2 diabetes mellitus (T2DM) is an independent risk factor of Alzheimer’s disease (AD), and populations with mild cognitive impairment (MCI) have high incidence to suffer from AD. Therefore, discerning who may be more vulnerable to MCI, among the increasing T2DM populations, is important for early intervention and eventually decreasing the prevalence rate of AD. This study was to explore whether the change of plasma β-amyloid (Aβ) could be a biomarker to distinguish MCI (T2DM-MCI) from non-MCI (T2DM-nMCI) in T2DM patients. Methods: Eight hundred fifty-two T2DM patients collected from five medical centers were assigned randomly to training and validation cohorts. Plasma Aβ, platelet glycogen synthase kinase-3β (GSK-3β), apolipoprotein E (ApoE) genotypes, and olfactory and cognitive functions were measured by ELISA, dot blot, RT-PCR, Connecticut Chemosensory Clinical Research Center (CCCRC) olfactory test based on the diluted butanol, and Minimum Mental State Examination (MMSE) test, respectively, and multivariate logistic regression analyses were applied. Results: Elevation of plasma Aβ1-42/Aβ1-40 is an independent risk factor of MCI in T2DM patients. Although using Aβ1-42/Aβ1-40 alone only reached an AUC of 0.631 for MCI diagnosis, addition of the elevated Aβ1-42/Aβ1-40 to our previous model (i.e., activated platelet GSK-3β, ApoE ε4 genotype, olfactory decline, and aging) significantly increased the discriminating efficiency of T2DM-MCI from T2DM-nMCI, with an AUC of 0.846 (95% CI: 0.794–0.897) to 0.869 (95% CI: 0.822–0.916) in the training cohort and an AUC of 0.848 (95% CI: 0.815–0.882) to 0.867 (95% CI: 0.835–0.899) in the validation cohort, respectively. Conclusion: A combination of the elevated plasma Aβ1-42/Aβ1-40 with activated platelet GSK-3β, ApoE ε4 genotype, olfactory decline, and aging could efficiently diagnose MCI in T2DM patients. Further longitudinal studies may consummate the model for early prediction of AD.
Collapse
Affiliation(s)
- Yanchao Liu
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Neurosurgery, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Shujuan Zhang
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Benrong He
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liangkai Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan Ke
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shi Zhao
- Department of Endocrinology, Central Hospital of Wuhan, Wuhan, China
| | - Yao Zhang
- Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wei
- Department of Endocrinology, Central Hospital of Wuhan, Wuhan, China
| | - Zhipeng Xu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zihui Xu
- Department of Endocrinology, Central Hospital of Wuhan, Wuhan, China
| | - Ying Yin
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen Mo
- Health Service Center of Jianghan District, Wuhan, China
| | - Yanni Li
- Health Service Center of Jianghan District, Wuhan, China
| | - Yang Gao
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shihong Li
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weijin Wang
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiling Yu
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongqin Wu
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guilin Pi
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Jiang
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingmin Deng
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Xiong
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiyang Lei
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Na Tian
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting He
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Sun
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuzhi Zhou
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Wang
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinwang Ye
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengzhu Li
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nan Hu
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guoda Song
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenju Peng
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenghong Zheng
- Department of Endocrinology, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Huaqiu Zhang
- Department of Neurosurgery, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Zhi Wang
- Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
7
|
Koychev I, Jansen K, Dette A, Shi L, Holling H. Blood-Based ATN Biomarkers of Alzheimer's Disease: A Meta-Analysis. J Alzheimers Dis 2021; 79:177-195. [PMID: 33252080 DOI: 10.3233/jad-200900] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The Amyloid Tau Neurodegeneration (ATN) framework was proposed to define the biological state underpinning Alzheimer's disease (AD). Blood-based biomarkers offer a scalable alternative to the costly and invasive currently available biomarkers. OBJECTIVE In this meta-analysis we sought to assess the diagnostic performance of plasma amyloid (Aβ40, Aβ42, Aβ42/40 ratio), tangle (p-tau181), and neurodegeneration (total tau [t-tau], neurofilament light [NfL]) biomarkers. METHODS Electronic databases were screened for studies reporting biomarker concentrations for AD and control cohorts. Biomarker performance was examined by random-effect meta-analyses based on the ratio between biomarker concentrations in patients and controls. RESULTS 83 studies published between 1996 and 2020 were included in the analyses. Aβ42/40 ratio as well as Aβ42 discriminated AD patients from controls when using novel platforms such as immunomagnetic reduction (IMR). We found significant differences in ptau-181 concentration for studies based on single molecule array (Simoa), but not for studies based on IMR or ELISA. T-tau was significantly different between AD patients and control in IMR and Simoa but not in ELISA-based studies. In contrast, NfL differentiated between groups across platforms. Exosome studies showed strong separation between patients and controls for Aβ42, t-tau, and p-tau181. CONCLUSION Currently available assays for sampling plasma ATN biomarkers appear to differentiate between AD patients and controls. Novel assay methodologies have given the field a significant boost for testing these biomarkers, such as IMR for Aβ, Simoa for p-tau181. Enriching samples through extracellular vesicles shows promise but requires further validation.
Collapse
Affiliation(s)
- Ivan Koychev
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Katrin Jansen
- Department of Psychology, University of Münster, Münster, Germany
| | - Alina Dette
- Department of Psychology, University of Münster, Münster, Germany
| | - Liu Shi
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Heinz Holling
- Department of Psychology, University of Münster, Münster, Germany
| |
Collapse
|
8
|
Hefner M, Baliga V, Amphay K, Ramos D, Hegde V. Cardiometabolic Modification of Amyloid Beta in Alzheimer's Disease Pathology. Front Aging Neurosci 2021; 13:721858. [PMID: 34497507 PMCID: PMC8419421 DOI: 10.3389/fnagi.2021.721858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 07/26/2021] [Indexed: 12/14/2022] Open
Abstract
In recent years, several studies have suggested that cardiometabolic disorders, such as diabetes, obesity, hypertension, and dyslipidemia, share strong connections with the onset of neurodegenerative disorders such as Parkinson's and Alzheimer's disease (AD). However, establishing a definitive link between medical disorders with coincident pathophysiologies is difficult due to etiological heterogeneity and underlying comorbidities. For this reason, amyloid β (Aβ), a physiological peptide derived from the sequential proteolysis of amyloid precursor protein (APP), serves as a crucial link that bridges the gap between cardiometabolic and neurodegenerative disorders. Aβ normally regulates neuronal synaptic function and repair; however, the intracellular accumulation of Aβ within the brain has been observed to play a critical role in AD pathology. A portion of Aβ is believed to originate from the brain itself and can readily cross the blood-brain barrier, while the rest resides in peripheral tissues that express APP required for Aβ generation such as the liver, pancreas, kidney, spleen, skin, and lungs. Consequently, numerous organs contribute to the body pool of total circulating Aβ, which can accumulate in the brain and facilitate neurodegeneration. Although the accumulation of Aβ corresponds with the onset of neurodegenerative disorders, the direct function of periphery born Aβ in AD pathophysiology is currently unknown. This review will highlight the contributions of individual cardiometabolic diseases including cardiovascular disease (CVD), type 2 diabetes (T2D), obesity, and non-alcoholic fatty liver disease (NAFLD) in elevating concentrations of circulating Aβ within the brain, as well as discuss the comorbid association of Aβ with AD pathology.
Collapse
Affiliation(s)
- Marleigh Hefner
- Obesity and Metabolic Health Laboratory, Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, United States
| | - Vineet Baliga
- College of Arts and Sciences, University of North Carolina, Chapel Hill, Chapel Hill, NC, United States
| | - Kailinn Amphay
- Obesity and Metabolic Health Laboratory, Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, United States
| | - Daniela Ramos
- Obesity and Metabolic Health Laboratory, Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, United States
| | - Vijay Hegde
- Obesity and Metabolic Health Laboratory, Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, United States
| |
Collapse
|
9
|
Nakai T, Yamada K, Mizoguchi H. Alzheimer's Disease Animal Models: Elucidation of Biomarkers and Therapeutic Approaches for Cognitive Impairment. Int J Mol Sci 2021; 22:5549. [PMID: 34074018 PMCID: PMC8197360 DOI: 10.3390/ijms22115549] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is an age-related and progressive neurodegenerative disorder. It is widely accepted that AD is mainly caused by the accumulation of extracellular amyloid β (Aβ) and intracellular neurofibrillary tau tangles. Aβ begins to accumulate years before the onset of cognitive impairment, suggesting that the benefit of currently available interventions would be greater if they were initiated in the early phases of AD. To understand the mechanisms of AD pathogenesis, various transgenic mouse models with an accelerated accumulation of Aβ and tau tangles have been developed. However, none of these models exhibit all pathologies present in human AD. To overcome these undesirable phenotypes, APP knock-in mice, which were presented with touchscreen-based tasks, were developed to better evaluate the efficacy of candidate therapeutics in mouse models of early-stage AD. This review assesses several AD mouse models from the aspect of biomarkers and cognitive impairment and discusses their potential as tools to provide novel AD therapeutic approaches.
Collapse
Affiliation(s)
- Tsuyoshi Nakai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan; (T.N.); (K.Y.)
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan; (T.N.); (K.Y.)
| | - Hiroyuki Mizoguchi
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan; (T.N.); (K.Y.)
- Medical Interactive Research and Academia Industry Collaboration Center, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
| |
Collapse
|
10
|
Deniz K, Ho CCG, Malphrus KG, Reddy JS, Nguyen T, Carnwath TP, Crook JE, Lucas JA, Graff-Radford NR, Carrasquillo MM, Ertekin-Taner N. Plasma Biomarkers of Alzheimer's Disease in African Americans. J Alzheimers Dis 2021; 79:323-334. [PMID: 33252078 PMCID: PMC7902984 DOI: 10.3233/jad-200828] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Background/Objective: The aim of this study was to determine if plasma concentrations of 5 surrogate markers of Alzheimer’s disease (AD) pathology and neuroinflammation are associated with disease status in African Americans. Methods: We evaluated 321 African Americans (159 AD, 162 controls) from the Florida Consortium for African-American Alzheimer’s Disease Studies (FCA3DS). Five plasma proteins reflecting AD neuropathology or inflammation (Aβ42, tau, IL6, IL10, TNFα) were tested for associations with AD, age, sex, APOE and MAPT genotypes, and for pairwise correlations. Results: Plasma tau levels were higher in AD when adjusted for biological and technical covariates. APOEɛ4 was associated with lower plasma Aβ42 and tau levels. Older age was associated with higher plasma Aβ42, tau, and TNFα. Females had lower IL10 levels. Inflammatory proteins had strong pairwise correlations amongst themselves and with Aβ42. Conclusion: We identified effects of demographic and genetic variants on five potential plasma biomarkers in African Americans. Plasma inflammatory biomarkers and Aβ42 may reflect correlated pathologies and elevated plasma tau may be a biomarker of AD in this population.
Collapse
Affiliation(s)
- Kaancan Deniz
- Mayo Clinic, Department of Neuroscience, Jacksonville, FL, USA
| | | | | | - Joseph S Reddy
- Mayo Clinic, Department of Health Science Research, Jacksonville, FL, USA
| | - Thuy Nguyen
- Mayo Clinic, Department of Neuroscience, Jacksonville, FL, USA
| | - Troy P Carnwath
- Mayo Clinic, Department of Neuroscience, Jacksonville, FL, USA
| | - Julia E Crook
- Mayo Clinic, Department of Health Science Research, Jacksonville, FL, USA
| | - John A Lucas
- Mayo Clinic, Department of Psychiatry and Psychology, Jacksonville, FL, USA
| | | | | | - Nilüfer Ertekin-Taner
- Mayo Clinic, Department of Neuroscience, Jacksonville, FL, USA.,Mayo Clinic, Department of Neurology, Jacksonville, FL, USA
| |
Collapse
|
11
|
Sung WH, Hung JT, Lu YJ, Cheng CM. Paper-Based Detection Device for Alzheimer's Disease-Detecting β-amyloid Peptides (1-42) in Human Plasma. Diagnostics (Basel) 2020; 10:E272. [PMID: 32365918 PMCID: PMC7277973 DOI: 10.3390/diagnostics10050272] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 12/19/2022] Open
Abstract
The diagnosis of Alzheimer's disease (AD) is frequently missed or delayed in clinical practice. To remedy this situation, we developed a screening, paper-based (P-ELISA) platform to detect β-amyloid peptide 1-42 (Aβ42) and provide rapid results using a small volume, easily accessible plasma sample instead of cerebrospinal fluid. The protocol outlined herein only requires 3 μL of sample per well and a short operating time (i.e., only 90 min). The detection limit of Aβ42 is 63.04 pg/mL in a buffer system. This P-ELISA-based approach can be used for early, preclinical stage AD screening, including screening for amnestic mild cognitive impairment (MCI) due to AD. It may also be used for treatment and stage monitoring purposes. The implementation of this approach may provide tremendous impact for an afflicted population and may well prompt additional and expanded efforts in both academic and commercial communities.
Collapse
Affiliation(s)
- Wei-Hsuan Sung
- Chang Gung Memorial Hospital Linkou Medical Center, Taoyuan 33305, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Jung-Tung Hung
- Institute of Stem Cell & Translational Cancer Research, Chang Gung Memorial Hospital Linkuo Medical Center, Taoyuan 33305, Taiwan;
| | - Yu-Jen Lu
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital Linkou Medical Center, Taoyuan 33305, Taiwan
| | - Chao-Min Cheng
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| |
Collapse
|
12
|
Lopez OL, Klunk WE, Mathis CA, Snitz BE, Chang Y, Tracy RP, Kuller LH. Relationship of amyloid-β1-42 in blood and brain amyloid: Ginkgo Evaluation of Memory Study. Brain Commun 2019; 2:fcz038. [PMID: 31998865 PMCID: PMC6976616 DOI: 10.1093/braincomms/fcz038] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/24/2019] [Accepted: 11/25/2019] [Indexed: 12/13/2022] Open
Abstract
A blood test that predicts the extent of amyloid plaques in the brain and risk of Alzheimer's disease would have important benefits for the early identification of higher risk of dementia and Alzheimer's disease and the evaluation of new preventative therapies. The goal of this study was to determine whether plasma levels of amyloid-β1-42, 1-40 and the amyloid-β1-42/1-40 ratio among participants in the Pittsburgh centre of the Ginkgo Evaluation of Memory Study were related to the extent of brain fibrillar amyloid plaques measured in 2009 using Pittsburgh compound-B PET imaging, hippocampal volume, cortical thickness in the temporal lobe and white matter lesions. There were 194 participants who had Pittsburgh compound-B measurements in 2009 with the mean age of 85 years; 96% were white and 60% men. Pittsburgh compound-B positivity was defined as a standardized uptake value ratio of ≥1.57. Amyloid-β in blood was measured using a sandwich enzyme-linked immunosorbent assay developed by Eli Lilly and modified at the University of Vermont. All participants were nondemented as of 2008 at the time of study close out. The study sample included 160 with blood samples drawn in 2000-02 and 133 from 2009 and also had brain amyloid measured in 2009. All blood samples were analysed at the same time in 2009. Plasma amyloid-β1-42 was inversely related to the percent Pittsburgh compound-B positive (standardized uptake value ratio ≥1.57), β -0.04, P = 0.005. Practically all participants who were apolipoprotein-E4 positive at older ages were also Pittsburgh compound-B positive for fibrillar amyloid. Among apolipoprotein-E4-negative participants, quartiles of amyloid-β1-42 were inversely related to Pittsburgh compound-B positivity. In multiple regression models, plasma amyloid-β1-42 measured in 2000-02 or 2009 were significantly and inversely related to Pittsburgh compound-B positivity as was the amyloid-β1-42/1-40 ratio. There was a 4-fold increase in the odds ratio for the presence of Pittsburgh compound-B positivity in the brain in 2009 for the first quartile of amyloid-β1-42 as compared with the fourth quartile in the multiple logistic model. This is one of the first longitudinal studies to evaluate the relationship between amyloid-β1-42 in the blood and the extent of brain amyloid deposition measured by PET imaging using Pittsburgh compound-B. Our findings showed that remote and recent low plasma amyloid-β1-42 levels were inversely associated with brain amyloid deposition in cognitively normal individuals. However, changes in plasma amyloid-β1-42 over time (8 years) were small and not related to the amount of Pittsburgh compound-B.
Collapse
Affiliation(s)
- Oscar L Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - William E Klunk
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Chester A Mathis
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Beth E Snitz
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Yuefang Chang
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Russell P Tracy
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Lewis H Kuller
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
13
|
Das BC, Dasgupta S, Ray SK. Potential therapeutic roles of retinoids for prevention of neuroinflammation and neurodegeneration in Alzheimer's disease. Neural Regen Res 2019; 14:1880-1892. [PMID: 31290437 PMCID: PMC6676868 DOI: 10.4103/1673-5374.259604] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 03/20/2019] [Indexed: 01/03/2023] Open
Abstract
All retinoids, which can be natural and synthetic, are chemically related to vitamin A. Both natural and synthetic retinoids use specific nuclear receptors such as retinoic acid receptors and retinoid X receptors to activate specific signaling pathways in the cells. Retinoic acid signaling is extremely important in the central nervous system. Impairment of retinoic acid signaling pathways causes severe pathological processes in the central nervous system, especially in the adult brain. Retinoids have major roles in neural patterning, differentiation, axon outgrowth in normal development, and function of the brain. Impaired retinoic acid signaling results in neuroinflammation, oxidative stress, mitochondrial malfunction, and neurodegeneration leading to progressive Alzheimer's disease, which is pathologically characterized by extra-neuronal accumulation of amyloid plaques (aggregated amyloid-beta) and intra-neurofibrillary tangles (hyperphosphorylated tau protein) in the temporal lobe of the brain. Alzheimer's disease is the most common cause of dementia and loss of memory in old adults. Inactive cholinergic neurotransmission is responsible for cognitive deficits in Alzheimer's disease patients. Deficiency or deprivation of retinoic acid in mice is associated with loss of spatial learning and memory. Retinoids inhibit expression of chemokines and neuroinflammatory cytokines in microglia and astrocytes, which are activated in Alzheimer's disease. Stimulation of retinoic acid receptors and retinoid X receptors slows down accumulation of amyloids, reduces neurodegeneration, and thereby prevents pathogenesis of Alzheimer's disease in mice. In this review, we described chemistry and biochemistry of some natural and synthetic retinoids and potentials of retinoids for prevention of neuroinflammation and neurodegeneration in Alzheimer's disease.
Collapse
Affiliation(s)
- Bhaskar C. Das
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Somsankar Dasgupta
- Department of Neuroscience and Regenerative Medicine, Institute of Molecular Medicine and Genetics, Augusta University, Augusta, GA, USA
| | - Swapan K. Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, USA
| |
Collapse
|
14
|
Wang J, Qiao F, Shang S, Li P, Chen C, Dang L, Jiang Y, Huo K, Deng M, Wang J, Qu Q. Elevation of Plasma Amyloid-β Level is More Significant in Early Stage of Cognitive Impairment: A Population-Based Cross-Sectional Study. J Alzheimers Dis 2019; 64:61-69. [PMID: 29865072 DOI: 10.3233/jad-180140] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Aggregation and deposition of amyloid-β (Aβ) in the brain is the main pathological change of Alzheimer's disease (AD). Decreased Aβ42 in the cerebrospinal fluid has been confirmed as a biomarker of AD; however, the relationship between plasma Aβ and cognitive impairment is currently unclear. OBJECTIVE The aim was to explore the relationship between plasma Aβ and cognitive impairment in a cross-sectional study. METHODS A total of 1,314 subjects (age above 40) from a village in the suburbs of Xi'an, China were enrolled between October 8, 2014 and March 30, 2015. A validated Chinese version of the Mini-Mental State Examination and neuropsychological battery were used to assess cognition. Levels of plasma Aβ42 and Aβ40 were tested using commercial enzyme-linked immunosorbent assay. Relationship of plasma Aβ and cognitive impairment was analyzed using logistic regression analysis. RESULTS Of the enrolled subjects, 1,180 (89.80%) had normal cognition, 85 (6.47%) had possible cognitive impairment and 49 (3.73%) had probable cognitive impairment. Logistic regression analysis showed that the Aβ42/Aβ40 ratio (OR = 4.042, 95% CI: 1.248-11.098, p = 0.012) and plasma Aβ42 (OR = 1.036, 95% CI: 1.003-1.071, p = 0.031) was higher in the possible cognitive impairment than that in the normal cognition group. Furthermore, the plasma Aβ42/Aβ40 ratio was higher in the possible cognitive impairment group than that in the probable cognitive impairment group (OR = 0.029, 95% CI: 0.002-0.450, p = 0.011). CONCLUSIONS Levels of plasma Aβ42 and Aβ42/Aβ40 ratio were elevated in patients with possible cognitive impairment, indicating that plasma Aβ42 and Aβ42/Aβ40 ratio increases may be more pronounced in early stage of cognitive impairment.
Collapse
Affiliation(s)
- Jin Wang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fan Qiao
- Weinan Central hospital, Shaanxi, China
| | - Suhang Shang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Pei Li
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chen Chen
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liangjun Dang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yu Jiang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Kang Huo
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meiying Deng
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jingyi Wang
- Huxian Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Qiumin Qu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
15
|
Kim Y, Yoo YK, Kim HY, Roh JH, Kim J, Baek S, Lee JC, Kim HJ, Chae MS, Jeong D, Park D, Lee S, Jang H, Kim K, Lee JH, Byun BH, Park SY, Ha JH, Lee KC, Cho WW, Kim JS, Koh JY, Lim SM, Hwang KS. Comparative analyses of plasma amyloid-β levels in heterogeneous and monomerized states by interdigitated microelectrode sensor system. SCIENCE ADVANCES 2019; 5:eaav1388. [PMID: 31001580 PMCID: PMC6469948 DOI: 10.1126/sciadv.aav1388] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 02/25/2019] [Indexed: 05/31/2023]
Abstract
Detection of amyloid-β (Aβ) aggregates contributes to the diagnosis of Alzheimer disease (AD). Plasma Aβ is deemed a less invasive and more accessible hallmark of AD, as Aβ can penetrate blood-brain barriers. However, correlations between biofluidic Aβ concentrations and AD progression has been tenuous. Here, we introduce a diagnostic technique that compares the heterogeneous and the monomerized states of Aβ in plasma. We used a small molecule, EPPS [4-(2-hydroxyethyl)-1-piperazinepropanesulfonic acid], to dissociate aggregated Aβ into monomers to enhance quantification accuracy. Subsequently, Aβ levels of EPPS-treated plasma were compared to those of untreated samples to minimize inter- and intraindividual variations. The interdigitated microelectrode sensor system was used to measure plasma Aβ levels on a scale of 0.1 pg/ml. The implementation of this self-standard blood test resulted in substantial distinctions between patients with AD and individuals with normal cognition (NC), with selectivity and sensitivity over 90%.
Collapse
Affiliation(s)
- YoungSoo Kim
- Integrated Science and Engineering Division, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Department of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Yong Kyoung Yoo
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Department of Electrical Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon-gu, Seoul 01897, Republic of Korea
| | - Hye Yun Kim
- Department of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Jee Hoon Roh
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Jinsik Kim
- Department of Medical Biotechnology, Dongguk University, 30 Pildong-ro 1-gil, Jung-gu, Seoul 04620, Republic of Korea
| | - Seungyeop Baek
- Department of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Department of Biotechnology, Yonsei University, 50 Yonsei-ro Seodaemun-gu, Seoul 03722, Republic of Korea
- Brain Science Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Jinny Claire Lee
- Integrated Science and Engineering Division, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Department of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Hye Jin Kim
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Myung-Sic Chae
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Dahye Jeong
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Dongsung Park
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Sejin Lee
- Department of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Brain Science Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - HoChung Jang
- Department of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Brain Science Institute, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Kyeonghwan Kim
- Department of Pharmacy, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
- Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, Republic of Korea
| | - Jeong Hoon Lee
- Department of Electrical Engineering, Kwangwoon University, 20 Kwangwoon-ro, Nowon-gu, Seoul 01897, Republic of Korea
| | - Byung Hyun Byun
- Department of Nuclear Medicine, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea
| | - Su Yeon Park
- Department of Neurology of Korea Cancer Center Hospital, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea
| | - Jeong Ho Ha
- Department of Neurology of Korea Cancer Center Hospital, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea
| | - Kyo Chul Lee
- Division of RI-Convergence Research, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea
| | - Won Woo Cho
- Cantis, Sangnok-gu, Ansan-si, Gyeonggi-do 15588, Republic of Korea
| | - Jae-Seung Kim
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Jae-Young Koh
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Sang Moo Lim
- Department of Neurology of Korea Cancer Center Hospital, Korea Institute of Radiological & Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea
| | - Kyo Seon Hwang
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| |
Collapse
|
16
|
Louis ED, Joyce JL, Cosentino S. Mind the gaps: What we don't know about cognitive impairment in essential tremor. Parkinsonism Relat Disord 2019; 63:10-19. [PMID: 30876840 DOI: 10.1016/j.parkreldis.2019.02.038] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 02/18/2019] [Accepted: 02/23/2019] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Although the hallmark feature of essential tremor (ET) is tremor, there is growing appreciation that cognitive impairment also occurs, including increased prevalence of mild cognitive impairment (MCI) and increased prevalence and incidence of dementia. With emerging knowledge of ET-cognitive impairment, come fundamental questions regarding its course, bases, predictors and clinical outcomes. Studies in the general population and in Parkinson's disease (PD), a related movement disorder, offer a starting point from which to begin filling these clinically important knowledge gaps. METHODS A PubMed search (June 2018) identified articles for this review. RESULTS Much of our knowledge of cognitive impairment in ET is of the static condition (e.g., prevalence of cognitive impairment in ET), with nearly no information on its bases, predictors and dynamics (i.e., course, and clinical outcomes). In PD, where such data have been published, rates of cognitive decline and conversion to MCI/dementia are higher than in the general population. Predictors of cognitive change in PD and the general population have also been identified, yet they only partially overlap one another. CONCLUSION The predictors and dynamics of cognitive impairment have been investigated fairly extensively in the general population, to a somewhat lesser extent in PD, and are emerging only now in ET. We suggest that longitudinal studies specific to ET are needed, and we outline variables to be considered in these investigations. Increased knowledge of ET-cognitive impairment will facilitate meaningful counseling of patients and their families.
Collapse
Affiliation(s)
- Elan D Louis
- Division of Movement Disorders, Department of Neurology, Yale School of Medicine, Yale University, New Haven, CT, USA; Department of Chronic Disease Epidemiology, Yale School of Public Health, Yale University, New Haven, CT, USA; Center for Neuroepidemiology and Clinical Neurological Research, Yale School of Medicine, Yale University, New Haven, CT, USA.
| | - Jillian L Joyce
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Stephanie Cosentino
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, USA; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| |
Collapse
|
17
|
Stock AJ, Kasus-Jacobi A, Pereira HA. The role of neutrophil granule proteins in neuroinflammation and Alzheimer's disease. J Neuroinflammation 2018; 15:240. [PMID: 30149799 PMCID: PMC6112130 DOI: 10.1186/s12974-018-1284-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 08/16/2018] [Indexed: 02/08/2023] Open
Abstract
Neutrophils are the innate immune system’s first line of defense. Neutrophils play a critical role in protecting the host against infectious pathogens, resolving sterile injuries, and mediating inflammatory responses. The granules of neutrophils and their constituent proteins are central to these functions. Although neutrophils may exert a protective role upon acute inflammatory conditions or insults, continued activity of neutrophils in chronic inflammatory diseases can contribute to tissue damage. Neutrophil granule proteins are involved in a number of chronic inflammatory conditions and diseases. However, the functions of these proteins in neuroinflammation and chronic neuroinflammatory diseases, including Alzheimer’s disease (AD), remain to be elucidated. In this review, we discuss recent findings from our lab and others that suggest possible functions for neutrophils and the neutrophil granule proteins, CAP37, neutrophil elastase, and cathepsin G, in neuroinflammation, with an emphasis on AD. These findings reveal that neutrophil granule proteins may exert both neuroprotective and neurotoxic effects. Further research should determine whether neutrophil granule proteins are valid targets for therapeutic interventions in chronic neuroinflammatory diseases.
Collapse
Affiliation(s)
- Amanda J Stock
- The Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd., BRC Rm 06B121, Baltimore, MD, 21224, USA.,Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 255, Oklahoma City, OK, 73117, USA
| | - Anne Kasus-Jacobi
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 255, Oklahoma City, OK, 73117, USA.,Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 255, Oklahoma City, OK, 73117, USA
| | - H Anne Pereira
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 255, Oklahoma City, OK, 73117, USA. .,Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 255, Oklahoma City, OK, 73117, USA. .,Department of Cell Biology, University of Oklahoma Health Sciences Center, 1105 N. Stonewall, Robert M. Bird Library, Rm 258, Oklahoma City, OK, 73117, USA. .,Department of Pathology, University of Oklahoma Health Sciences Center, 1105 N. Stonewall, Robert M. Bird Library, Rm 258, Oklahoma City, OK, 73117, USA.
| |
Collapse
|
18
|
Effect of Chlorogenic Acid Intake on Cognitive Function in the Elderly: A Pilot Study. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:8608497. [PMID: 29707036 PMCID: PMC5863287 DOI: 10.1155/2018/8608497] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 12/08/2017] [Accepted: 01/24/2018] [Indexed: 11/18/2022]
Abstract
Objective To evaluate the effect of chlorogenic acids (CGAs) intake on cognitive function. Methods In this pilot study, the Cogstate and CNS Vital Signs test batteries were used to evaluate cognitive function in 8 healthy elderly men and women complaining of subjective memory loss after a 6-month intake of a test beverage containing 330 mg of CGAs just before bedtime. Results After a 6-month CGA intake period, significant improvement was observed in the One Back Test of the Cogstate, the Shifting Attention Test, and Finger Tapping Test as well as in the composite memory, verbal memory, complex attention, cognitive flexibility, executive function, and motor speed domains of the CNS Vital Signs test battery. Conclusion A 6-month intake of CGAs may improve attentional, executive, and memory functions in the elderly with complaints of subjective memory loss.
Collapse
|
19
|
Boada M, Anaya F, Ortiz P, Olazarán J, Shua-Haim JR, Obisesan TO, Hernández I, Muñoz J, Buendia M, Alegret M, Lafuente A, Tárraga L, Núñez L, Torres M, Grifols JR, Ferrer I, Lopez OL, Páez A. Efficacy and Safety of Plasma Exchange with 5% Albumin to Modify Cerebrospinal Fluid and Plasma Amyloid-β Concentrations and Cognition Outcomes in Alzheimer's Disease Patients: A Multicenter, Randomized, Controlled Clinical Trial. J Alzheimers Dis 2018; 56:129-143. [PMID: 27911295 PMCID: PMC5240541 DOI: 10.3233/jad-160565] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: Studies conducted in animal models and humans suggest the presence of a dynamic equilibrium of amyloid-β (Aβ) peptide between cerebrospinal fluid (CSF) and plasma compartments. Objective: To determine whether plasma exchange (PE) with albumin replacement was able to modify Aβ concentrations in CSF and plasma as well as to improve cognition in patients with mild-moderate Alzheimer’s disease (AD). Methods: In a multicenter, randomized, patient- and rater-blind, controlled, parallel-group, phase II study, 42 AD patients were assigned (1 : 1) to PE treatment or control (sham) groups. Treated patients received a maximum of 18 PE with 5% albumin (Albutein®, Grifols) with three different schedules: two PE/weekly (three weeks), one PE/weekly (six weeks), and one PE/bi- weekly (12 weeks), plus a six-month follow-up period. Plasma and CSF Aβ1–40 and Aβ1–42 levels, as well as cognitive, functional, and behavioral measures were determined. Results: CSF Aβ1–42 levels after the last PE compared to baseline were marginally higher in PE-treated group versus controls (adjusted means of variation: 75.3 versus –45.5 pg/mL; 95% CI: –19.8, 170.5 versus 135.1, 44.2; p = 0.072). Plasma Aβ1–42 levels were lower in the PE-treated group after each treatment period (p < 0.05). Plasma Aβ1–40 levels showed a saw-tooth pattern variation associated with PE. PE-treated patients scored better in the Boston Naming Test and Semantic Verbal Fluency (p < 0.05) throughout the study. Neuropsychiatric Inventory scores were higher in controls during the PE phase (p < 0.05). Conclusion: PE with human albumin modified CSF and plasma Aβ1–42 levels. Patients treated with PE showed improvement in memory and language functions, which persisted after PE was discontinued.
Collapse
Affiliation(s)
- Mercè Boada
- Memory Clinic and Research Center of Fundació ACE, Institut Catalá de Neurociències Aplicades, Barcelona, Spain.,Neurology Service, Hospital General Universitari Vall d'Hebron, Barcelona, Spain
| | - Fernando Anaya
- Nephrology Service, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | | - Javier Olazarán
- Neurology Service, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Joshua R Shua-Haim
- Alzheimer's Research Corporation, Mid Atlantic Geriatric Association, Manchester, NJ, USA
| | - Thomas O Obisesan
- Department of Internal Medicine, Howard University, Washington, DC, USA
| | - Isabel Hernández
- Memory Clinic and Research Center of Fundació ACE, Institut Catalá de Neurociències Aplicades, Barcelona, Spain
| | - Joan Muñoz
- Banc de Sang i Teixits, Barcelona, Spain
| | - Mar Buendia
- Memory Clinic and Research Center of Fundació ACE, Institut Catalá de Neurociències Aplicades, Barcelona, Spain
| | - Montserrat Alegret
- Memory Clinic and Research Center of Fundació ACE, Institut Catalá de Neurociències Aplicades, Barcelona, Spain
| | - Asunción Lafuente
- Memory Clinic and Research Center of Fundació ACE, Institut Catalá de Neurociències Aplicades, Barcelona, Spain
| | - Lluís Tárraga
- Memory Clinic and Research Center of Fundació ACE, Institut Catalá de Neurociències Aplicades, Barcelona, Spain
| | - Laura Núñez
- Clinical Trials Department, Instituto Grifols S.A., Barcelona, Spain
| | - Mireia Torres
- Clinical Trials Department, Instituto Grifols S.A., Barcelona, Spain
| | | | - Isidre Ferrer
- Institut de Neuropatologia, Hospital Universitario Bellvitge, Barcelona, Spain
| | - Oscar L Lopez
- Departments of Neurology and Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Antonio Páez
- Clinical Trials Department, Instituto Grifols S.A., Barcelona, Spain
| |
Collapse
|
20
|
Slachevsky A, Guzmán-Martínez L, Delgado C, Reyes P, Farías GA, Muñoz-Neira C, Bravo E, Farías M, Flores P, Garrido C, Becker JT, López OL, Maccioni RB. Tau Platelets Correlate with Regional Brain Atrophy in Patients with Alzheimer's Disease. J Alzheimers Dis 2018; 55:1595-1603. [PMID: 27911301 DOI: 10.3233/jad-160652] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Intracellular neurofibrillary tangles are part of the core pathology of Alzheimer's disease (AD), which are mainly composed of hyperphosphorylated tau protein. OBJECTIVES The purpose of this study is to determine whether high molecular weight (HMW) or low molecular weight (LMW) tau protein levels, as well as the ratio HMW/LMW, present in platelets correlates with brain magnetic resonance imaging (MRI) structural changes in normal and cognitively impaired subjects. METHODS We examined 53 AD patients and 37 cognitively normal subjects recruited from two Memory Clinics at the Universidad de Chile. Tau levels in platelets were determined by immunoreactivity and the MRI scans were analyzed using voxel-based morphometry in 41 AD patients. RESULTS The HMW/LMW tau ratio was statistically different between controls and AD patients, and no associations were noted between HMW or LMW tau and MRI structures. In a multivariate analysis controlled for age and education level, the HMW/LMW tau ratio was associated with reduced volume in the left medial and right anterior cingulate gyri, right cerebellum, right thalamus (pulvinar), left frontal cortex, and right parahippocampal region. CONCLUSIONS This exploratory study showed that HMW/LMW tau ratio is significantly higher in AD patients than control subjects, and that it is associated with specific brain regions atrophy. Determination of peripheral markers of AD pathology can help understanding the pathophysiology of neurodegeneration in AD.
Collapse
Affiliation(s)
- Andrea Slachevsky
- Physiopathology Department, ICBM and East Neuroscience Department, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Cognitive Neurology and Dementia Unit, Neurological Sciences Department, Faculty of Medicine, Universidad de Chile and Neurology Department, Hospital del Salvador, Santiago, Chile.,Gerosciences Center for Brain Health and Metabolism, Santiago, Chile.,CIAE, Center for Advanced Research in Education, Universidad de Chile, Santiago, Chile
| | | | - Carolina Delgado
- Neurology and Neurosurgery Department, Clinical Hospital, Universidad de Chile, Santiago, Chile
| | - Pablo Reyes
- Physiopathology Department, ICBM and East Neuroscience Department, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Gonzalo A Farías
- International Center for Biomedicine, Santiago, Chile.,Neurology and Neurosurgery Department, Clinical Hospital, Universidad de Chile, Santiago, Chile
| | - Carlos Muñoz-Neira
- Cognitive Neurology and Dementia Unit, Neurological Sciences Department, Faculty of Medicine, Universidad de Chile and Neurology Department, Hospital del Salvador, Santiago, Chile
| | - Eduardo Bravo
- Neuroradiology Department, Institute of Neurosurgery Asenjo, Santiago, Chile
| | - Mauricio Farías
- Neuroradiology Department, Institute of Neurosurgery Asenjo, Santiago, Chile
| | - Patricia Flores
- Cognitive Neurology and Dementia Unit, Neurological Sciences Department, Faculty of Medicine, Universidad de Chile and Neurology Department, Hospital del Salvador, Santiago, Chile.,Neurology Unit, Department of Medicine, Clínica Alemana-Universidad del Desarollo, Santiago, Chile
| | - Cristian Garrido
- Neurology and Neurosurgery Department, Clinical Hospital, Universidad de Chile, Santiago, Chile
| | - James T Becker
- Department of Psychiatry, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA.,Department of Psychology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Oscar L López
- Department of Psychiatry, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA.,Department of Neurology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Ricardo B Maccioni
- International Center for Biomedicine, Santiago, Chile.,Faculty of Sciences, University of Chile, Santiago, Chile
| |
Collapse
|
21
|
Crane A, Brubaker WD, Johansson JU, Trigunaite A, Ceballos J, Bradt B, Glavis-Bloom C, Wallace TL, Tenner AJ, Rogers J. Peripheral complement interactions with amyloid β peptide in Alzheimer's disease: 2. Relationship to amyloid β immunotherapy. Alzheimers Dement 2018; 14:243-252. [PMID: 28755839 PMCID: PMC5881571 DOI: 10.1016/j.jalz.2017.04.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/27/2017] [Accepted: 04/30/2017] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Our previous studies have shown that amyloid β peptide (Aβ) is subject to complement-mediated clearance from the peripheral circulation, and that this mechanism is deficient in Alzheimer's disease. The mechanism should be enhanced by Aβ antibodies that form immune complexes (ICs) with Aβ, and therefore may be relevant to current Aβ immunotherapy approaches. METHODS Multidisciplinary methods were employed to demonstrate enhanced complement-mediated capture of Aβ antibody immune complexes compared with Aβ alone in both erythrocytes and THP1-derived macrophages. RESULTS Aβ antibodies dramatically increased complement activation and opsonization of Aβ, followed by commensurately enhanced Aβ capture by human erythrocytes and macrophages. These in vitro findings were consistent with enhanced peripheral clearance of intravenously administered Aβ antibody immune complexes in nonhuman primates. DISCUSSION Together with our previous results, showing significant Alzheimer's disease deficits in peripheral Aβ clearance, the present findings strongly suggest that peripheral mechanisms should not be ignored as contributors to the effects of Aβ immunotherapy.
Collapse
Affiliation(s)
- Andrés Crane
- Biosciences Division, SRI International, Menlo Park, CA, USA
| | | | | | | | | | - Bonnie Bradt
- Biosciences Division, SRI International, Menlo Park, CA, USA
| | | | - Tanya L Wallace
- Biosciences Division, SRI International, Menlo Park, CA, USA
| | - Andrea J Tenner
- Departments of Molecular Biology and Biochemistry and Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
| | - Joseph Rogers
- Biosciences Division, SRI International, Menlo Park, CA, USA.
| |
Collapse
|
22
|
Ishijima S, Baba H, Maeshima H, Shimano T, Inoue M, Suzuki T, Arai H. Glucocorticoid may influence amyloid β metabolism in patients with depression. Psychiatry Res 2018; 259:191-196. [PMID: 29073556 DOI: 10.1016/j.psychres.2017.10.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 09/25/2017] [Accepted: 10/02/2017] [Indexed: 11/19/2022]
Abstract
Epidemiological studies have demonstrated that depression may be a risk factor for Alzheimer's disease (AD); however, the biological mechanisms of the transition from depression to AD are still not clear. Changes of amyloid β protein (Aβ) metabolism and increased glucocorticoid (GC) levels have been found in both depression and AD. Moreover, several studies in animal models have demonstrated that GC administration changes Aβ metabolism. To reveal whether GC affects amyloid metabolism in patients with depression, we evaluated serum levels of Aβ40, Aβ42 and cortisol at admission in 187 inpatients with major depressive disorder (MDD) and 224 healthy comparisons. Additionally, we re-evaluated the serum levels of Aβs in 27 patients with MDD 1 year later. The results of multiple regression analyses revealed that serum cortisol and Aβ levels are not correlated at the time of admission. However, serum cortisol levels at admission correlated with serum Aβ42 levels and Aβ40/Aβ42 ratio 1 year later. These findings suggest that increased cortisol in patients with MDD may influence the metabolism of Aβ over prolonged periods of time.
Collapse
Affiliation(s)
- Satoko Ishijima
- Department of Psychiatry & Behavioral Science, Juntendo Graduate School of Medicine, Tokyo, Japan; Juntendo University Mood Disorder Project (JUMP), Department of Psychiatry, Juntendo Koshigaya Hospital, Saitama, Japan
| | - Hajime Baba
- Department of Psychiatry & Behavioral Science, Juntendo Graduate School of Medicine, Tokyo, Japan; Juntendo University Mood Disorder Project (JUMP), Department of Psychiatry, Juntendo Koshigaya Hospital, Saitama, Japan.
| | - Hitoshi Maeshima
- Juntendo University Mood Disorder Project (JUMP), Department of Psychiatry, Juntendo Koshigaya Hospital, Saitama, Japan
| | - Takahisa Shimano
- Juntendo University Mood Disorder Project (JUMP), Department of Psychiatry, Juntendo Koshigaya Hospital, Saitama, Japan
| | - Megumi Inoue
- Juntendo University Mood Disorder Project (JUMP), Department of Psychiatry, Juntendo Koshigaya Hospital, Saitama, Japan
| | - Toshihito Suzuki
- Department of Psychiatry & Behavioral Science, Juntendo Graduate School of Medicine, Tokyo, Japan; Juntendo University Mood Disorder Project (JUMP), Department of Psychiatry, Juntendo Koshigaya Hospital, Saitama, Japan
| | - Heii Arai
- Department of Psychiatry & Behavioral Science, Juntendo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
23
|
Ma F, Du H. Novel deoxyvasicinone derivatives as potent multitarget-directed ligands for the treatment of Alzheimer's disease: Design, synthesis, and biological evaluation. Eur J Med Chem 2017; 140:118-127. [DOI: 10.1016/j.ejmech.2017.09.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/03/2017] [Accepted: 09/05/2017] [Indexed: 12/31/2022]
|
24
|
Chen GF, Xu TH, Yan Y, Zhou YR, Jiang Y, Melcher K, Xu HE. Amyloid beta: structure, biology and structure-based therapeutic development. Acta Pharmacol Sin 2017; 38:1205-1235. [PMID: 28713158 PMCID: PMC5589967 DOI: 10.1038/aps.2017.28] [Citation(s) in RCA: 1020] [Impact Index Per Article: 145.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 03/02/2017] [Indexed: 12/12/2022] Open
Abstract
Amyloid beta peptide (Aβ) is produced through the proteolytic processing of a transmembrane protein, amyloid precursor protein (APP), by β- and γ-secretases. Aβ accumulation in the brain is proposed to be an early toxic event in the pathogenesis of Alzheimer's disease, which is the most common form of dementia associated with plaques and tangles in the brain. Currently, it is unclear what the physiological and pathological forms of Aβ are and by what mechanism Aβ causes dementia. Moreover, there are no efficient drugs to stop or reverse the progression of Alzheimer's disease. In this paper, we review the structures, biological functions, and neurotoxicity role of Aβ. We also discuss the potential receptors that interact with Aβ and mediate Aβ intake, clearance, and metabolism. Additionally, we summarize the therapeutic developments and recent advances of different strategies for treating Alzheimer's disease. Finally, we will report on the progress in searching for novel, potentially effective agents as well as selected promising strategies for the treatment of Alzheimer's disease. These prospects include agents acting on Aβ, its receptors and tau protein, such as small molecules, vaccines and antibodies against Aβ; inhibitors or modulators of β- and γ-secretase; Aβ-degrading proteases; tau protein inhibitors and vaccines; amyloid dyes and microRNAs.
Collapse
Affiliation(s)
- Guo-Fang Chen
- VARI-SIMM Center, Center for Structure and Function of Drug Targets, CAS-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ting-Hai Xu
- VARI-SIMM Center, Center for Structure and Function of Drug Targets, CAS-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yan Yan
- VARI-SIMM Center, Center for Structure and Function of Drug Targets, CAS-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yu-Ren Zhou
- VARI-SIMM Center, Center for Structure and Function of Drug Targets, CAS-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yi Jiang
- VARI-SIMM Center, Center for Structure and Function of Drug Targets, CAS-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Karsten Melcher
- Laboratory of Structural Sciences, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - H Eric Xu
- VARI-SIMM Center, Center for Structure and Function of Drug Targets, CAS-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Laboratory of Structural Sciences, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| |
Collapse
|
25
|
Peripheral complement interactions with amyloid β peptide: Erythrocyte clearance mechanisms. Alzheimers Dement 2017; 13:1397-1409. [PMID: 28475854 DOI: 10.1016/j.jalz.2017.03.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 02/28/2017] [Accepted: 03/27/2017] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Although amyloid β peptide (Aβ) is cleared from the brain to cerebrospinal fluid and the peripheral circulation, mechanisms for its removal from blood remain unresolved. Primates have uniquely evolved a highly effective peripheral clearance mechanism for pathogens, immune adherence, in which erythrocyte complement receptor 1 (CR1) plays a major role. METHODS Multidisciplinary methods were used to demonstrate immune adherence capture of Aβ by erythrocytes and its deficiency in Alzheimer's disease (AD). RESULTS Aβ was shown to be subject to immune adherence at every step in the pathway. Aβ dose-dependently activated serum complement. Complement-opsonized Aβ was captured by erythrocytes via CR1. Erythrocytes, Aβ, and hepatic Kupffer cells were colocalized in the human liver. Significant deficits in erythrocyte Aβ levels were found in AD and mild cognitive impairment patients. DISCUSSION CR1 polymorphisms elevate AD risk, and >80% of human CR1 is vested in erythrocytes to subserve immune adherence. The present results suggest that this pathway is pathophysiologically relevant in AD.
Collapse
|
26
|
Li W, Xu Z, Xu B, Chan CY, Lin X, Wang Y, Chen G, Wang Z, Yuan Q, Zhu G, Sun H, Wu W, Shi P. Investigation of the Subcellular Neurotoxicity of Amyloid-β Using a Device Integrating Microfluidic Perfusion and Chemotactic Guidance. Adv Healthc Mater 2017; 6. [PMID: 28121396 DOI: 10.1002/adhm.201600895] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 11/28/2016] [Indexed: 11/10/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with the histopathological hallmark of extracellular accumulation of amyloid-β (Aβ) peptide in brain senile plaques. Though many studies have shown the neural toxicity from various forms of Aβ peptides, the subcellular mechanisms of Aβ peptide are still not well understood, partially due to the technical challenges of isolating axons or dendrites from the cell body for localized investigation. In this study, the subcellular toxicity and localization of Aβ peptides are investigated by utilizing a microfluidic compartmentalized device, which combines physical restriction and chemotactic guidance to enable the isolation of axons and dendrites for localized pharmacological studies. It is found that Aβ peptides induced neuronal death is mostly resulted from Aβ treatment at cell body or axonal processes, but not at dendritic neurites. Simply applying Aβ to axons alone induces significant hyperactive spiking activity. Dynamic transport of Aβ aggregates is only observed between axon terminal and cell body. In addition to differential cellular uptake, more Aβ-peptide secretion is detected significantly from axons than from dendritic side. These results clearly demonstrate the existence of a localized mechanism in Aβ-induced neurotoxicity, and can potentially benefit the development of new therapeutic strategies for AD.
Collapse
Affiliation(s)
- Wei Li
- Department of Mechanical and Biomedical Engineering; City University of Hong Kong; 83 Tat Chee Ave Kowloon Hong Kong SAR 999077 China
| | - Zhen Xu
- Department of Mechanical and Biomedical Engineering; City University of Hong Kong; 83 Tat Chee Ave Kowloon Hong Kong SAR 999077 China
| | - Bingzhe Xu
- Department of Mechanical and Biomedical Engineering; City University of Hong Kong; 83 Tat Chee Ave Kowloon Hong Kong SAR 999077 China
| | - Chung Yuen Chan
- Department of Mechanical and Biomedical Engineering; City University of Hong Kong; 83 Tat Chee Ave Kowloon Hong Kong SAR 999077 China
| | - Xudong Lin
- Department of Mechanical and Biomedical Engineering; City University of Hong Kong; 83 Tat Chee Ave Kowloon Hong Kong SAR 999077 China
| | - Ying Wang
- Department of Mechanical and Biomedical Engineering; City University of Hong Kong; 83 Tat Chee Ave Kowloon Hong Kong SAR 999077 China
| | - Ganchao Chen
- Department of Biology and Chemistry; City University of Hong Kong; 83 Tat Chee Ave Kowloon Hong Kong SAR 999077 China
| | - Zhigang Wang
- Department of Biology and Chemistry; City University of Hong Kong; 83 Tat Chee Ave Kowloon Hong Kong SAR 999077 China
| | - Qiuju Yuan
- School of Chinese Medicine; Faculty of Science; The Chinese University of Hong Kong; Shatin, Hong Kong SAR 999077 China
| | - Guangyu Zhu
- Department of Biology and Chemistry; City University of Hong Kong; 83 Tat Chee Ave Kowloon Hong Kong SAR 999077 China
| | - Hongyan Sun
- Department of Biology and Chemistry; City University of Hong Kong; 83 Tat Chee Ave Kowloon Hong Kong SAR 999077 China
| | - Wutian Wu
- Department of Anatomy; The University of Hong Kong; 21 Sassoon Road Hong Kong SAR 999077 China
| | - Peng Shi
- Department of Mechanical and Biomedical Engineering; City University of Hong Kong; 83 Tat Chee Ave Kowloon Hong Kong SAR 999077 China
- Shenzhen Research Institute; City University of Hong Kong; Shenzhen 518057 P. R. China
| |
Collapse
|
27
|
Bode DC, Baker MD, Viles JH. Ion Channel Formation by Amyloid-β42 Oligomers but Not Amyloid-β40 in Cellular Membranes. J Biol Chem 2016; 292:1404-1413. [PMID: 27927987 DOI: 10.1074/jbc.m116.762526] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/24/2016] [Indexed: 12/23/2022] Open
Abstract
A central hallmark of Alzheimer's disease is the presence of extracellular amyloid plaques chiefly consisting of amyloid-β (Aβ) peptides in the brain interstitium. Aβ largely exists in two isoforms, 40 and 42 amino acids long, but a large body of evidence points to Aβ(1-42) rather than Aβ(1-40) as the cytotoxic form. One proposed mechanism by which Aβ exerts toxicity is the formation of ion channel pores that disrupt intracellular Ca2+ homeostasis. However, previous studies using membrane mimetics have not identified any notable difference in the channel forming properties between Aβ(1-40) and Aβ(1-42). Here, we tested whether a more physiological environment, membranes excised from HEK293 cells of neuronal origin, would reveal differences in the relative channel forming ability of monomeric, oligomeric, and fibrillar forms of both Aβ(1-40) and Aβ(1-42). Aβ preparations were characterized with transmission electron microscopy and thioflavin T fluorescence. Aβ was then exposed to the extracellular face of excised membranes, and transmembrane currents were monitored using patch clamp. Our data indicated that Aβ(1-42) assemblies in oligomeric preparations form voltage-independent, non-selective ion channels. In contrast, Aβ(1-40) oligomers, fibers, and monomers did not form channels. Ion channel conductance results suggested that Aβ(1-42) oligomers, but not monomers and fibers, formed three distinct pore structures with 1.7-, 2.1-, and 2.4-nm pore diameters. Our findings demonstrate that only Aβ(1-42) contains unique structural features that facilitate membrane insertion and channel formation, now aligning ion channel formation with the differential neurotoxic effect of Aβ(1-40) and Aβ(1-42) in Alzheimer's disease.
Collapse
Affiliation(s)
- David C Bode
- From the School of Biological and Chemical Sciences, Queen Mary, University of London, Mile End Road, London E1 4NS, United Kingdom and
| | - Mark D Baker
- the Blizard Institute, Centre for Neuroscience and Trauma, Barts and The London School of Medicine and Dentistry, Queen Mary, University of London, 4 Newark Street, London E1 2AT, United Kingdom
| | - John H Viles
- From the School of Biological and Chemical Sciences, Queen Mary, University of London, Mile End Road, London E1 4NS, United Kingdom and
| |
Collapse
|
28
|
Martins IJ. The Role of Clinical Proteomics, Lipidomics, and Genomics in the Diagnosis of Alzheimer's Disease. Proteomes 2016; 4:proteomes4020014. [PMID: 28248224 PMCID: PMC5217345 DOI: 10.3390/proteomes4020014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 03/24/2016] [Accepted: 03/28/2016] [Indexed: 02/07/2023] Open
Abstract
The early diagnosis of Alzheimer’s disease (AD) has become important to the reversal and treatment of neurodegeneration, which may be relevant to premature brain aging that is associated with chronic disease progression. Clinical proteomics allows the detection of various proteins in fluids such as the urine, plasma, and cerebrospinal fluid for the diagnosis of AD. Interest in lipidomics has accelerated with plasma testing for various lipid biomarkers that may with clinical proteomics provide a more reproducible diagnosis for early brain aging that is connected to other chronic diseases. The combination of proteomics with lipidomics may decrease the biological variability between studies and provide reproducible results that detect a community’s susceptibility to AD. The diagnosis of chronic disease associated with AD that now involves genomics may provide increased sensitivity to avoid inadvertent errors related to plasma versus cerebrospinal fluid testing by proteomics and lipidomics that identify new disease biomarkers in body fluids, cells, and tissues. The diagnosis of AD by various plasma biomarkers with clinical proteomics may now require the involvement of lipidomics and genomics to provide interpretation of proteomic results from various laboratories around the world.
Collapse
Affiliation(s)
- Ian James Martins
- School of Medical Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup 6027, Australia.
| |
Collapse
|
29
|
Wang Z, Li W, Wang Y, Li X, Huang L, Li X. Design, synthesis and evaluation of clioquinol–ebselen hybrids as multi-target-directed ligands against Alzheimer's disease. RSC Adv 2016. [DOI: 10.1039/c5ra26797h] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
A novel series of compounds obtained by fusing the metal-chelating agent clioquinol and the antioxidant ebselen were designed, synthesized and evaluated as multi-target-directed ligands against Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Zhiren Wang
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- China
| | - Wenrui Li
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- China
| | - Yali Wang
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- China
| | - Xiruo Li
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- China
| | - Ling Huang
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- China
| | - Xingshu Li
- School of Pharmaceutical Sciences
- Sun Yat-sen University
- Guangzhou
- China
| |
Collapse
|
30
|
[Clinically validated molecular biomarkers of neurodegenerative dementia]. DER NERVENARZT 2015; 85:1372-81. [PMID: 25331214 DOI: 10.1007/s00115-014-4086-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
As cerebrospinal fluid-based neurochemical dementia diagnostics (CSF-NDD) has now been validated at the S3 evidence level, the German Association for Psychiatry, Psychotherapy and Psychosomatics (DGPPN) and the German Society for Neurology (DGN) recommend CSF-NDD in the recent joint dementia guidelines for improved early and differential diagnostics of multigenic (sporadic) Alzheimer's dementia (AD). The CSF-NDD also provides a predictive diagnosis of incipient AD for high-risk patients when they are still in the prodromal stage of mild cognitive impairment (MCI) but as no (secondary) preventive therapy of AD is currently available, the use of CSF-NDD for the predictive molecular diagnosis of AD is not recommended in the neuropsychiatry guidelines (http://www.DGPPN.de). However, molecular diagnostics of preclinical AD by CSF-NDD and/or [18F]-amyloid positron emission tomography (PET) has meanwhile gained high clinical relevance for therapeutic clinical research, as this novel clinical model allows systematic screening for promising (secondary) preventive therapy options. Moreover, it has now become apparent that blood-based neurochemical diagnostics of preclinical and early AD will be possible by means of various formats of multiplex assays. However, so far promising blood assays have not been consistently validated by independent research groups and in contrast to CSF-NDD a blood-based diagnosis of AD is not yet available.
Collapse
|
31
|
Donohue MC, Moghadam SH, Roe AD, Sun CK, Edland SD, Thomas RG, Petersen RC, Sano M, Galasko D, Aisen PS, Rissman RA. Longitudinal plasma amyloid beta in Alzheimer's disease clinical trials. Alzheimers Dement 2015; 11:1069-79. [PMID: 25301682 PMCID: PMC4387108 DOI: 10.1016/j.jalz.2014.07.156] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/13/2014] [Accepted: 07/05/2014] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Little is known about the utility of plasma amyloid beta (Aβ) in clinical trials of Alzheimer's disease (AD). METHODS We analyzed longitudinal plasma samples from two large multicenter clinical trials: (1) donezepil and vitamin E in mild cognitive impairment (n = 405, 24 months) and (2) simvastatin in mild to moderate AD (n = 225, 18 months). RESULTS Baseline plasma Aβ was not related to cognitive or clinical progression. We observed a decrease in plasma Aβ40 and 42 among apolipoprotein E epsilon 4 (APOE ε4) carriers relative to noncarriers in the mild cognitive impairment trial. Patients treated with simvastatin showed a significant increase in Aβ compared with placebo. We found significant storage time effects and considerable plate-to-plate variation. DISCUSSION We found no support for the utility of plasma Aβ as a prognostic factor or correlate of cognitive change. Analysis of stored specimens requires careful standardization and experimental design, but plasma Aβ may prove useful in pharmacodynamic studies of antiamyloid drugs.
Collapse
Affiliation(s)
- Michael C Donohue
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, University of California San Diego, School of Medicine, San Diego, CA, USA; Department of Family Preventive Medicine, University of California San Diego, School of Medicine, San Diego, CA, USA
| | - Setareh H Moghadam
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, University of California San Diego, School of Medicine, San Diego, CA, USA
| | - Allyson D Roe
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, University of California San Diego, School of Medicine, San Diego, CA, USA
| | - Chung-Kai Sun
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, University of California San Diego, School of Medicine, San Diego, CA, USA
| | - Steven D Edland
- Department of Family Preventive Medicine, University of California San Diego, School of Medicine, San Diego, CA, USA
| | - Ronald G Thomas
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, University of California San Diego, School of Medicine, San Diego, CA, USA; Department of Family Preventive Medicine, University of California San Diego, School of Medicine, San Diego, CA, USA
| | - Ronald C Petersen
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, University of California San Diego, School of Medicine, San Diego, CA, USA; Department of Neurology, Mayo Clinic Alzheimer's Disease Research Center, Department of Health Sciences Mayo Clinic College of Medicine, Research, Rochester, MN, USA
| | - Mary Sano
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, University of California San Diego, School of Medicine, San Diego, CA, USA; Mount Sinai School of Medicine and James J. Peters Veterans Association Medical Center, Bronx, NY, USA
| | - Douglas Galasko
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, University of California San Diego, School of Medicine, San Diego, CA, USA
| | - Paul S Aisen
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, University of California San Diego, School of Medicine, San Diego, CA, USA
| | - Robert A Rissman
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, University of California San Diego, School of Medicine, San Diego, CA, USA.
| |
Collapse
|
32
|
Shanthi KB, Krishnan S, Rani P. A systematic review and meta-analysis of plasma amyloid 1-42 and tau as biomarkers for Alzheimer's disease. SAGE Open Med 2015; 3:2050312115598250. [PMID: 26770797 PMCID: PMC4679337 DOI: 10.1177/2050312115598250] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 06/27/2015] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Amyloid 1-42 (Aβ42) and tau in cerebrospinal fluid are currently used as markers for diagnosis of Alzheimer's disease. Conflicting reports exist regarding their plasma levels in Alzheimer's disease patients. A meta-analysis was performed to statistically validate the use of plasma Aβ42 and tau as biomarkers for Alzheimer's disease. METHODS Different databases were searched using the search key: (amyloid OR amyloid1-42 OR Aβ42) AND (tau OR total tau) AND plasma AND (alzheimer's OR alzheimer's disease), and for databases not accepting boolean search, records were retrieved using the search key: plasma + amyloid + tau + alzheimer's. A total of 1880 articles for Aβ42 and 1508 articles for tau were shortlisted. The abstracts were screened, and 69 articles reporting plasma Aβ42 levels and 6 articles reporting plasma tau were identified. After exclusion, 25 studies reporting plasma Aβ42 and 6 studies reporting total tau were analysed in Review Manager version 5.2 using weighted mean difference method, and the bias between studies was assessed using the funnel plot. RESULTS Plasma Aβ42 and tau did not vary significantly between Alzheimer's disease patients and controls. The funnel plot showed that there was no bias between studies for Aβ42, while possible bias existed for tau due to availability of limited studies. CONCLUSION This analysis pinpoints that plasma Aβ42 and tau could not serve as reliable markers independently for diagnosis of Alzheimer's disease and a cohort study with age, sex and apolipoprotein E correction is warranted for their possible use as Alzheimer's disease markers.
Collapse
Affiliation(s)
| | - Sreeram Krishnan
- Department of Biotechnology, PSG College of Technology, Coimbatore, India
| | - P Rani
- Department of Biotechnology, PSG College of Technology, Coimbatore, India
| |
Collapse
|
33
|
Schupf N, Lee A, Park N, Dang LH, Pang D, Yale A, Oh DKT, Krinsky-McHale SJ, Jenkins EC, Luchsinger JA, Zigman WB, Silverman W, Tycko B, Kisselev S, Clark L, Lee JH. Candidate genes for Alzheimer's disease are associated with individual differences in plasma levels of beta amyloid peptides in adults with Down syndrome. Neurobiol Aging 2015; 36:2907.e1-10. [PMID: 26166206 DOI: 10.1016/j.neurobiolaging.2015.06.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 06/08/2015] [Accepted: 06/14/2015] [Indexed: 01/08/2023]
Abstract
We examined the contribution of candidates genes for Alzheimer's disease (AD) to individual differences in levels of beta amyloid peptides in adults with Down syndrom, a population at high risk for AD. Participants were 254 non-demented adults with Down syndrome, 30-78 years of age. Genomic deoxyribonucleic acid was genotyped using an Illumina GoldenGate custom array. We used linear regression to examine differences in levels of Aβ peptides associated with the number of risk alleles, adjusting for age, sex, level of intellectual disability, race and/or ethnicity, and the presence of the APOE ε4 allele. For Aβ42 levels, the strongest gene-wise association was found for a single nucleotide polymorphism (SNP) on CAHLM1; for Aβ40 levels, the strongest gene-wise associations were found for SNPs in IDE and SOD1, while the strongest gene-wise associations with levels of the Aβ42/Aβ40 ratio were found for SNPs in SORCS1. Broadly classified, variants in these genes may influence amyloid precursor protein processing (CALHM1, IDE), vesicular trafficking (SORCS1), and response to oxidative stress (SOD1).
Collapse
Affiliation(s)
- Nicole Schupf
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA; G.H. Sergievsky Center, New York, NY, USA; Department of Epidemiology, Columbia University Medical Center, New York, NY, USA; Department of Psychiatry, Columbia University Medical Center, New York, NY, USA.
| | - Annie Lee
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA
| | - Naeun Park
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA
| | - Lam-Ha Dang
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA
| | - Deborah Pang
- Department of Psychology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Alexander Yale
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - David Kyung-Taek Oh
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Sharon J Krinsky-McHale
- Department of Psychology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Edmund C Jenkins
- Department of Human Genetics, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - José A Luchsinger
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Warren B Zigman
- Department of Psychology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Wayne Silverman
- Kennedy Krieger Institute and Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Benjamin Tycko
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Sergey Kisselev
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA
| | - Lorraine Clark
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA
| | - Joseph H Lee
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA; G.H. Sergievsky Center, New York, NY, USA; Department of Epidemiology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
34
|
Hallé M, Tribout-Jover P, Lanteigne AM, Boulais J, St-Jean JR, Jodoin R, Girouard MP, Constantin F, Migneault A, Renaud F, Didierlaurent AM, Mallett CP, Burkhart D, Pilorget A, Palmantier R, Larocque D. Methods to monitor monocytes-mediated amyloid-beta uptake and phagocytosis in the context of adjuvanted immunotherapies. J Immunol Methods 2015; 424:64-79. [PMID: 26002154 DOI: 10.1016/j.jim.2015.05.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 04/11/2015] [Accepted: 05/06/2015] [Indexed: 12/14/2022]
Abstract
Antibody-mediated capture of amyloid-beta (Aβ) in peripheral blood was identified as an attractive strategy to eliminate cerebral toxic amyloid in Alzheimer's disease (AD) patients and murine models. Alternatively, defective capacity of peripheral monocytes to engulf Aβ was reported in individuals with AD. In this report, we developed different approaches to investigate cellular uptake and phagocytosis of Aβ, and to examine how two immunological devices--an immunostimulatory Adjuvant System and different amyloid specific antibodies--may affect these biological events. Between one and thirteen months of age, APPswe X PS1.M146V (TASTPM) AD model mice had decreasing concentrations of Aβ in their plasma. In contrast, the proportion of blood monocytes containing Aβ tended to increase with age. Importantly, the TLR-agonist containing Adjuvant System AS01B primed monocytes to promote de novo Aβ uptake capacity, particularly in the presence of anti-Aβ antibodies. Biochemical experiments demonstrated that cells achieved Aβ uptake and internalization followed by Aβ degradation via mechanisms that required effective actin polymerization and proteolytic enzymes such as insulin-degrading enzyme. We further demonstrated that both Aβ-specific monoclonal antibodies and plasma from Aβ-immunized mice enhanced the phagocytosis of 1 μm Aβ-coated particles. Together, our data highlight a new biomarker testing to follow amyloid clearance within the blood and a mechanism of Aβ uptake by peripheral monocytes in the context of active or passive immunization, and emphasize on novel approaches to investigate this phenomenon.
Collapse
Affiliation(s)
- Maxime Hallé
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8; Neuroscience Laboratory, 2705, Boulevard Laurier, T-2-50, Department of Molecular Medicine, Centre Hospitalier Universitaire de Québec Research Center, Université Laval, Quebec City, Quebec, Canada, G1V 4G2
| | | | | | - Jonathan Boulais
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8
| | - Julien R St-Jean
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8
| | - Rachel Jodoin
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8
| | | | - Florin Constantin
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8
| | - Annik Migneault
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8
| | - Frédéric Renaud
- GSK Vaccines, Rue de l'Institut 89, B-1330 Rixensart, Belgium
| | | | - Corey P Mallett
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8
| | - David Burkhart
- GSK Vaccines, 553 Old Corvallis Road, Hamilton, MT 59840, USA
| | - Anthony Pilorget
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8
| | - Rémi Palmantier
- GSK Vaccines, Rue de l'Institut 89, B-1330 Rixensart, Belgium
| | - Daniel Larocque
- GSK Vaccines, 525 Boulevard Cartier Ouest, Laval, Quebec, Canada, H7V 3S8.
| |
Collapse
|
35
|
Abstract
Alzheimer's disease (AD), the most common form of dementia in western societies, is a pathologically and clinically heterogeneous disease with a strong genetic component. The recent advances in high-throughput genome technologies allowing for the rapid analysis of millions of polymorphisms in thousands of subjects has significantly advanced our understanding of the genomic underpinnings of AD susceptibility. During the last 5 years, genome-wide association and whole-exome- and whole-genome sequencing studies have mapped more than 20 disease-associated loci, providing insights into the molecular pathways involved in AD pathogenesis and hinting at potential novel therapeutic targets. This review article summarizes the challenges and opportunities of when using genomic information for the diagnosis and prognosis of AD.
Collapse
Affiliation(s)
- Christiane Reitz
- Sergievsly Center/Taub Institute/Dept. of Neurology, Columbia University, 630 W 168th Street, Rm 19-308, New York, NY 10032, phone: (212) 305-0865, fax: (212) 305-2391
| |
Collapse
|
36
|
Zhao Z, Zhu L, Bu X, Ma H, Yang S, Yang Y, Hu Z. Label-free detection of Alzheimer’s disease through the ADP3 peptoid recognizing the serum amyloid-beta42 peptide. Chem Commun (Camb) 2015; 51:718-21. [DOI: 10.1039/c4cc07037b] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Surface plasmon resonance imaging in combination with the ADP3 peptoid was used to identify Alzheimer’s disease through detecting amyloid-beta42 in the serum.
Collapse
Affiliation(s)
- Zijian Zhao
- National Center for Nanoscience and Technology
- Beijing
- P.R. China
| | - Ling Zhu
- National Center for Nanoscience and Technology
- Beijing
- P.R. China
| | - Xiangli Bu
- National Center for Nanoscience and Technology
- Beijing
- P.R. China
| | - Huailei Ma
- National Center for Nanoscience and Technology
- Beijing
- P.R. China
| | - Shu Yang
- National Center for Nanoscience and Technology
- Beijing
- P.R. China
| | - Yanlian Yang
- National Center for Nanoscience and Technology
- Beijing
- P.R. China
| | - Zhiyuan Hu
- National Center for Nanoscience and Technology
- Beijing
- P.R. China
| |
Collapse
|
37
|
Abstract
The complement component receptor 1 gene (CR1), which encodes a type-I transmembrane glycoprotein, has recently been identified as one of the most important risk genes for late-onset Alzheimer's disease (LOAD). In this article, we reviewed the recent evidence concerning the role of CR1 in LOAD. First, we introduced the structure, localization and physiological function of CR1 in humans. Afterward, we summarized the relation of CR1 polymorphisms with LOAD risk. Finally, we discussed the possible impact of CR1 on the pathogenesis of AD including amyloid-β pathology, tauopathy, immune dysfunction and glial-mediated neuroinflammation. We hope that a more comprehensive understanding of the role that CR1 played in AD may lead to the development of novel therapeutics for the prevention and treatment of AD.
Collapse
|
38
|
A blood-based predictor for neocortical Aβ burden in Alzheimer's disease: results from the AIBL study. Mol Psychiatry 2014; 19:519-26. [PMID: 23628985 DOI: 10.1038/mp.2013.40] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 01/30/2013] [Accepted: 03/06/2013] [Indexed: 12/28/2022]
Abstract
Dementia is a global epidemic with Alzheimer's disease (AD) being the leading cause. Early identification of patients at risk of developing AD is now becoming an international priority. Neocortical Aβ (extracellular β-amyloid) burden (NAB), as assessed by positron emission tomography (PET), represents one such marker for early identification. These scans are expensive and are not widely available, thus, there is a need for cheaper and more widely accessible alternatives. Addressing this need, a blood biomarker-based signature having efficacy for the prediction of NAB and which can be easily adapted for population screening is described. Blood data (176 analytes measured in plasma) and Pittsburgh Compound B (PiB)-PET measurements from 273 participants from the Australian Imaging, Biomarkers and Lifestyle (AIBL) study were utilised. Univariate analysis was conducted to assess the difference of plasma measures between high and low NAB groups, and cross-validated machine-learning models were generated for predicting NAB. These models were applied to 817 non-imaged AIBL subjects and 82 subjects from the Alzheimer's Disease Neuroimaging Initiative (ADNI) for validation. Five analytes showed significant difference between subjects with high compared to low NAB. A machine-learning model (based on nine markers) achieved sensitivity and specificity of 80 and 82%, respectively, for predicting NAB. Validation using the ADNI cohort yielded similar results (sensitivity 79% and specificity 76%). These results show that a panel of blood-based biomarkers is able to accurately predict NAB, supporting the hypothesis for a relationship between a blood-based signature and Aβ accumulation, therefore, providing a platform for developing a population-based screen.
Collapse
|
39
|
Qiu WWQ, Lai A, Mon T, Mwamburi M, Taylor W, Rosenzweig J, Kowall N, Stern R, Zhu H, Steffens DC. Angiotensin converting enzyme inhibitors and Alzheimer disease in the presence of the apolipoprotein E4 allele. Am J Geriatr Psychiatry 2014; 22:177-85. [PMID: 23567418 PMCID: PMC3873370 DOI: 10.1016/j.jagp.2012.08.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 07/11/2012] [Accepted: 08/29/2012] [Indexed: 11/21/2022]
Abstract
OBJECTIVE The effect of angiotensin converting enzyme (ACE) inhibitors on Alzheimer disease (AD) remains unclear, with conflicting results reported. We studied the interaction of the Apolipoprotein E (ApoE) genotype and ACE inhibitors on AD. METHODS This was a cross-sectional study of homebound elderly with an AD diagnosis and documentation of medications taken. ApoE genotype was determined. RESULTS A total of 355 subjects with status on ApoE alleles and cognitive diagnoses were studied. The average age (mean ± SD) of this population was 73.3 ± 8.3 years old, and 73% were female. Cross-sectionally, there was no difference in the number of AD cases between ApoE4 carriers and ApoE4 non-carriers or between ACE inhibitor users and non-users in the homebound elderly. ApoE4 carriers treated with ACE inhibitors, however, had more diagnoses of AD compared with those who did not have the treatment (28% versus 6%, p = 0.01) or ApoE4 non-carriers treated with an ACE inhibitor (28% versus 10%, p = 0.03). ACE inhibitor use was associated with AD diagnosis only in the presence of an E4 allele. Using multivariate logistic regression analysis, we found that in diagnosed AD cases there was a significant interaction between ApoE4 and ACE inhibitor use (odds ratio: 20.85; 95% confidence interval: 3.08-140.95; p = 0.002) after adjusting for age, sex, ethnicity, and education. CONCLUSION The effects of ACE inhibitors on AD may be different depending on ApoE genotype. A prospective study is needed to determine whether ACE inhibitor use accelerates or poorly delays AD development in ApoE4 carriers compared with ApoE4 non-carriers.
Collapse
Affiliation(s)
- Wendy Wei Qiao Qiu
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA; Department of Psychiatry, Boston University School of Medicine, Boston, MA; Department of Alzheimer Disease Center, Boston University School of Medicine, Boston, MA.
| | - Angela Lai
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA
| | - Timothy Mon
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA
| | - Mkaya Mwamburi
- Department of Public Health and Community Medicine, Tufts University, Boston, MA
| | - Warren Taylor
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC
| | - James Rosenzweig
- Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Neil Kowall
- Department of Alzheimer Disease Center, Boston University School of Medicine, Boston, MA
| | - Robert Stern
- Department of Alzheimer Disease Center, Boston University School of Medicine, Boston, MA
| | - Haihao Zhu
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA
| | - David C Steffens
- Department of Psychiatry, University of Connecticut Health Center, Farmington, CT
| |
Collapse
|
40
|
Abstract
Dementia is currently diagnosed based on clinical symptoms and signs, but significant brain damage has already occurred by the time a clinical diagnosis of dementia is made, and it is increasingly recognized that this may be too late for any effective intervention. It would therefore be of great public health and preventive value to define a variety of biomarkers that could permit early detection of persons at a higher risk for developing dementia, and specifically dementia due to Alzheimer's disease. Nevertheless, for the purpose of large-scale screening, circulating biomarkers are more appropriate because they are less invasive than lumbar puncture, less costly than brain amyloid imaging and can be easily assessed repeatedly in a primary care clinic setting. In this brief review we will review a number of candidate molecules implicated as possible predictors of dementia risk. These candidates include markers of vascular injury, metabolic and inflammatory states, amyloid and tau pathway markers, measures of neural degeneration and repair efforts, and other molecules that might contribute to anatomical and functional changes characteristic of dementia and Alzheimer's disease.
Collapse
Affiliation(s)
- Galit Weinstein
- Department of Neurology, Boston University School of Medicine, B602, 72 East Concord Street, Boston, MA 02118, USA
- Framingham Heart Study, 73 Mount Wayte Avenue, Framingham, MA 01702, USA
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, B602, 72 East Concord Street, Boston, MA 02118, USA
- Framingham Heart Study, 73 Mount Wayte Avenue, Framingham, MA 01702, USA
| |
Collapse
|
41
|
Du X, Wang Z, Zheng Y, Li H, Ni J, Liu Q. Inhibitory Effect of Selenoprotein P on Cu+/Cu2+-Induced Aβ42 Aggregation and Toxicity. Inorg Chem 2014; 53:1672-8. [DOI: 10.1021/ic4028282] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Xiubo Du
- Department of Marine Biology, Shenzhen
Key Laboratory of Marine Biotechnology and Ecology, Shenzhen University, Shenzhen 518060, China
| | - Zhi Wang
- College
of Life Sciences, Shenzhen Key Laboratory of Microbial Genetic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Youbiao Zheng
- Department of Marine Biology, Shenzhen
Key Laboratory of Marine Biotechnology and Ecology, Shenzhen University, Shenzhen 518060, China
| | - Haiping Li
- College
of Life Sciences, Shenzhen Key Laboratory of Microbial Genetic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Jiazuan Ni
- College
of Life Sciences, Shenzhen Key Laboratory of Microbial Genetic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Qiong Liu
- Department of Marine Biology, Shenzhen
Key Laboratory of Marine Biotechnology and Ecology, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
42
|
Reitz C, Mayeux R. Alzheimer disease: epidemiology, diagnostic criteria, risk factors and biomarkers. Biochem Pharmacol 2014; 88:640-51. [PMID: 24398425 DOI: 10.1016/j.bcp.2013.12.024] [Citation(s) in RCA: 766] [Impact Index Per Article: 76.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 12/20/2013] [Accepted: 12/20/2013] [Indexed: 12/20/2022]
Abstract
The global prevalence of dementia is as high as 24 million, and has been predicted to quadruple by the year 2050. In the US alone, Alzheimer disease (AD) - the most frequent cause of dementia characterized by a progressive decline in cognitive function in particular the memory domain - causes estimated health-care costs of $ 172 billion per year. Key neuropathological hallmarks of the AD brain are diffuse and neuritic extracellular amyloid plaques - often surrounded by dystrophic neurites - and intracellular neurofibrillary tangles. These pathological changes are frequently accompanied by reactive microgliosis and loss of neurons, white matter and synapses. The etiological mechanisms underlying these neuropathological changes remain unclear, but are probably caused by both environmental and genetic factors. In this review article, we provide an overview of the epidemiology of AD, review the biomarkers that may be used for risk assessment and in diagnosis, and give suggestions for future research.
Collapse
Affiliation(s)
- Christiane Reitz
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, United States; Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, United States; Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, United States
| | - Richard Mayeux
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, United States; Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, United States; Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, United States; Department of Epidemiology, Joseph P. Mailman School of Public Health, Columbia University, New York, NY, United States; Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY, United States.
| |
Collapse
|
43
|
Lee SL, Thomas P, Fenech M. Extracellular amyloid beta 42 causes necrosis, inhibition of nuclear division, and mitotic disruption under both folate deficient and folate replete conditions as measured by the cytokinesis-block micronucleus cytome assay. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2014; 55:1-14. [PMID: 24038346 DOI: 10.1002/em.21811] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 07/17/2013] [Accepted: 07/19/2013] [Indexed: 06/02/2023]
Abstract
Alzheimer's disease is associated with accumulation of extracellular beta amyloid peptide 42 (Aβ42) which may induce DNA damage and reduce cellular regenerative potential. These effects may be exacerbated under conditions of folate deficiency. The aim of this study was to investigate whether extracellular Aβ42 induces DNA damage and cell death in human peripheral lymphocytes and whether there is an interactive effect between extracellular Aβ42 and folic acid status. Peripheral blood lymphocytes were cultured in medium under conditions of both low and high folate (20 and 200 nM, respectively) and challenged with either Aβ42 or the physiologically normal form Aβ40 (both at 5, 10, 15 µM). Genome stability and cytotoxicity events were investigated using the cytokinesis-block micronucleus cytome (CBMN-cyt) assay. Outcome measures scored included the nuclear division index (NDI), necrosis, apoptosis, binucleated cells with micronuclei (MN), nucleoplasmic bridges (NPB), and nuclear buds (NBUD) and abnormally shaped nuclei (circular, (CIR) and horse-shoe, (HS) that may be indicative of mitotic disruption. Folic acid deficiency significantly reduced NDI (P < 0.001) and increased all the DNA damage biomarkers (MN, NPB, NBUD, HS, CIR), (P < 0.001). In contrast, exposure to Aβ40 had no impact on CBMN cytome biomarkers but Aβ42 significantly reduced NDI (P < 0.01), increased necrosis (P < 0.05) and frequency of cells with circular nuclei (P < 0.01). There was no evidence of an interaction between Aβ42 and folic acid with respect to CBMN cytome biomarkers. Extracellular Aβ42 appears to have cytotoxic and cytostatic effects but its effect on chromosomal instability appears to be small relative to folate deficiency.
Collapse
Affiliation(s)
- Sau Lai Lee
- Commonwealth Scientific and Industrial Research Organisation, (Animal, Food and Health Sciences), Adelaide, South Australia, 5000; Discipline of Physiology, School of Medical Sciences, The University of Adelaide, Adelaide, South Australia, 5005
| | | | | |
Collapse
|
44
|
Davanipour Z, Tseng CC, Lee PJ, Markides KS, Sobel E. Severe Cognitive Dysfunction and Occupational Extremely Low Frequency Magnetic Field Exposure among Elderly Mexican Americans. ACTA ACUST UNITED AC 2013; 4:1641-1662. [PMID: 24839595 PMCID: PMC4020120 DOI: 10.9734/bjmmr/2014/7317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Aims This report is the first study of the possible relationship between extremely low frequency (50–60 Hz, ELF) magnetic field (MF) exposure and severe cognitive dysfunction. Earlier studies investigated the relationships between MF occupational exposure and Alzheimer’s disease (AD) or dementia. These studies had mixed results, depending upon whether the diagnosis of AD or dementia was performed by experts and upon the methodology used to classify MF exposure. Study Design Population-based case-control. Place and Duration of Study Neurology and Preventive Medicine, Keck School of Medicine, University of Southern California, 2 years. Methodology The study population consisted of 3050 Mexican Americans, aged 65+, enrolled in Phase 1 of the Hispanic Established Population for the Epidemiologic Study of the Elderly (H-EPESE) study. Mini-Mental State Exam (MMSE) results, primary occupational history, and other data were collected. Severe cognitive dysfunction was defined as an MMSE score below 10. The MF exposure methodology developed and used in earlier studies was used. Results Univariate odds ratios (OR) were 3.4 (P< .03; 95% CI: 1.3–8.9) for high and 1.7 (P=.27; 95% CI: 0.7–4.1) for medium or high (M/H) MF occupations. In multivariate main effects models, the results were similar. When interaction terms were allowed in the models, the interactions between M/H or high occupational MF exposure and smoking history or age group were statistically significant, depending upon whether two (65–74, 75+) or three (65–74, 75–84, 85+) age groups were considered, respectively. When the analyses were limited to subjects aged 75+, the interactions between M/H or high MF occupations and a positive smoking history were statistically significant. Conclusion The results of this study indicate that working in an occupation with high or M/H MF exposure may increase the risk of severe cognitive dysfunction. Smoking and older age may increase the deleterious effect of MF exposure.
Collapse
Affiliation(s)
- Zoreh Davanipour
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Chiu-Chen Tseng
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Pey-Jiuan Lee
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Kyriakos S Markides
- Department of Preventive Medicine and Community Health, University of Texas Medical Branch, Galveston, Texas 77555-1153, USA
| | - Eugene Sobel
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA ; Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
45
|
Hampel H, Lista S, Teipel SJ, Garaci F, Nisticò R, Blennow K, Zetterberg H, Bertram L, Duyckaerts C, Bakardjian H, Drzezga A, Colliot O, Epelbaum S, Broich K, Lehéricy S, Brice A, Khachaturian ZS, Aisen PS, Dubois B. Perspective on future role of biological markers in clinical therapy trials of Alzheimer's disease: a long-range point of view beyond 2020. Biochem Pharmacol 2013; 88:426-49. [PMID: 24275164 DOI: 10.1016/j.bcp.2013.11.009] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 11/13/2013] [Accepted: 11/13/2013] [Indexed: 10/26/2022]
Abstract
Recent advances in understanding the molecular mechanisms underlying various paths toward the pathogenesis of Alzheimer's disease (AD) has begun to provide new insight for interventions to modify disease progression. The evolving knowledge gained from multidisciplinary basic research has begun to identify new concepts for treatments and distinct classes of therapeutic targets; as well as putative disease-modifying compounds that are now being tested in clinical trials. There is a mounting consensus that such disease modifying compounds and/or interventions are more likely to be effectively administered as early as possible in the cascade of pathogenic processes preceding and underlying the clinical expression of AD. The budding sentiment is that "treatments" need to be applied before various molecular mechanisms converge into an irreversible pathway leading to morphological, metabolic and functional alterations that characterize the pathophysiology of AD. In light of this, biological indicators of pathophysiological mechanisms are desired to chart and detect AD throughout the asymptomatic early molecular stages into the prodromal and early dementia phase. A major conceptual development in the clinical AD research field was the recent proposal of new diagnostic criteria, which specifically incorporate the use of biomarkers as defining criteria for preclinical stages of AD. This paradigm shift in AD definition, conceptualization, operationalization, detection and diagnosis represents novel fundamental opportunities for the modification of interventional trial designs. This perspective summarizes not only present knowledge regarding biological markers but also unresolved questions on the status of surrogate indicators for detection of the disease in asymptomatic people and diagnosis of AD.
Collapse
Affiliation(s)
- Harald Hampel
- Université Pierre et Marie Curie, Département de Neurologie, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Pavillon François Lhermitte, Hôpital de la Salpêtrière, Paris, France.
| | - Simone Lista
- Department of Psychiatry, Psychotherapy and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Halle/Saale, Germany.
| | - Stefan J Teipel
- Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany; German Center for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Rostock, Germany
| | - Francesco Garaci
- Department of Diagnostic Imaging, Molecular Imaging, Interventional Radiology, and Radiotherapy, University of Rome "Tor Vergata", Rome, Italy; IRCCS San Raffaele Pisana, Rome and San Raffaele Cassino, Cassino, Italy
| | - Robert Nisticò
- Department of Physiology and Pharmacology, University of Rome "La Sapienza", Rome, Italy; IRCSS Santa Lucia Foundation, Rome, Italy
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; University College London Institute of Neurology, Queen Square, London, UK
| | - Lars Bertram
- Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Charles Duyckaerts
- Laboratoire de Neuropathologie Raymond-Escourolle, Groupe Hospitalier Pitié-Salpêtrière, AP-HP, Paris, France
| | - Hovagim Bakardjian
- IM2A - Institute of Memory and Alzheimer's Disease, Paris, France; IHU-A-ICM - Paris Institute of Translational Neurosciences Pitié-Salpêtrière University Hospital, Paris, France
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital of Cologne, Cologne, Germany
| | - Olivier Colliot
- Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moelle Épinière, UMR-S975 Paris, France; Inserm, U975, Paris, France; CNRS, UMR 7225, Paris, France; ICM - Institut du Cerveau et de la Moelle Épinière, Paris, France; INRIA, Aramis Team, Centre de Recherche Paris-Rocquencourt, France
| | - Stéphane Epelbaum
- Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié Salpêtrière, Paris, France; Université Pierre et Marie Curie, Paris, France
| | - Karl Broich
- Federal Institute of Drugs and Medical Devices (BfArM), Bonn, Germany
| | - Stéphane Lehéricy
- IHU-A-ICM - Paris Institute of Translational Neurosciences Pitié-Salpêtrière University Hospital, Paris, France; Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moelle Épinière, UMR-S975 Paris, France; Inserm, U975, Paris, France; CNRS, UMR 7225, Paris, France; ICM - Institut du Cerveau et de la Moelle Épinière, Paris, France
| | - Alexis Brice
- Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moelle Épinière, UMR-S975 Paris, France; Inserm, U975, Paris, France; CNRS, UMR 7225, Paris, France; ICM - Institut du Cerveau et de la Moelle Épinière, Paris, France; AP-HP, Hôpital de la Salpêtrière, Département de Génétique et Cytogénétique, Paris, France
| | | | - Paul S Aisen
- Department of Neurosciences, University of California, San Diego, San Diego, CA, USA
| | - Bruno Dubois
- Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié Salpêtrière, Paris, France; Université Pierre et Marie Curie, Paris, France
| |
Collapse
|
46
|
Butterfield DA, Swomley AM, Sultana R. Amyloid β-peptide (1-42)-induced oxidative stress in Alzheimer disease: importance in disease pathogenesis and progression. Antioxid Redox Signal 2013; 19:823-35. [PMID: 23249141 PMCID: PMC3749710 DOI: 10.1089/ars.2012.5027] [Citation(s) in RCA: 380] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 12/05/2012] [Accepted: 12/17/2012] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE Alzheimer disease (AD) is an age-related neurodegenerative disease. AD is characterized by progressive cognitive impairment. One of the main histopathological hallmarks of AD brain is the presence of senile plaques (SPs) and another is elevated oxidative stress. The main component of SPs is amyloid beta-peptide (Aβ) that is derived from the proteolytic cleavage of amyloid precursor protein. RECENT ADVANCES Recent studies are consistent with the notion that methionine present at 35 position of Aβ is critical to Aβ-induced oxidative stress and neurotoxicity. Further, we also discuss the signatures of oxidatively modified brain proteins, identified using redox proteomics approaches, during the progression of AD. CRITICAL ISSUES The exact relationships of the specifically oxidatively modified proteins in AD pathogenesis require additional investigation. FUTURE DIRECTIONS Further studies are needed to address whether the therapies directed toward brain oxidative stress and oxidatively modified key brain proteins might help delay or prevent the progression of AD.
Collapse
Affiliation(s)
- D Allan Butterfield
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506-0055, USA.
| | | | | |
Collapse
|
47
|
Rembach A, Ryan TM, Roberts BR, Doecke JD, Wilson WJ, Watt AD, Barnham KJ, Masters CL. Progress towards a consensus on biomarkers for Alzheimer’s disease: a review of peripheral analytes. Biomark Med 2013; 7:641-62. [DOI: 10.2217/bmm.13.59] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of dementia in the elderly population and attempts to develop therapies have been unsuccessful because there is no means to target an effective therapeutic window. CNS biomarkers are insightful but impractical for high-throughput population-based screening. Therefore, a peripheral, blood-based biomarker for AD would significantly improve early diagnosis, potentially enable presymptomatic detection and facilitate effective targeting of disease-modifying treatments. The various constituents of blood, including plasma, platelets and cellular fractions, are now being systematically explored as a pool of putative peripheral biomarkers for AD. In this review we cover some less known peripheral biomarkers and highlight the latest developments for their clinical application.
Collapse
Affiliation(s)
- Alan Rembach
- The Mental Health Research Institute, The University of Melbourne, Kenneth Myer Building, 30 Royal Parade, Parkville, Victoria, 3010, Australia.
| | - Tim M Ryan
- The Mental Health Research Institute, The University of Melbourne, Kenneth Myer Building, 30 Royal Parade, Parkville, Victoria, 3010, Australia
| | - Blaine R Roberts
- The Mental Health Research Institute, The University of Melbourne, Kenneth Myer Building, 30 Royal Parade, Parkville, Victoria, 3010, Australia
| | - James D Doecke
- The Australian e-Health Research Centre, Herston, Queensland, 4029, Australia
- CSIRO Preventative Health National Research Flagship, North Ryde, New South Wales, 2113, Australia
| | - William J Wilson
- CSIRO Preventative Health National Research Flagship, North Ryde, New South Wales, 2113, Australia
| | - Andrew D Watt
- The Mental Health Research Institute, The University of Melbourne, Kenneth Myer Building, 30 Royal Parade, Parkville, Victoria, 3010, Australia
| | - Kevin J Barnham
- The Mental Health Research Institute, The University of Melbourne, Kenneth Myer Building, 30 Royal Parade, Parkville, Victoria, 3010, Australia
| | - Colin L Masters
- The Mental Health Research Institute, The University of Melbourne, Kenneth Myer Building, 30 Royal Parade, Parkville, Victoria, 3010, Australia
| |
Collapse
|
48
|
Maftei M, Thurm F, Schnack C, Tumani H, Otto M, Elbert T, Kolassa IT, Przybylski M, Manea M, von Arnim CAF. Increased levels of antigen-bound β-amyloid autoantibodies in serum and cerebrospinal fluid of Alzheimer's disease patients. PLoS One 2013; 8:e68996. [PMID: 23874844 PMCID: PMC3715516 DOI: 10.1371/journal.pone.0068996] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Accepted: 06/04/2013] [Indexed: 12/11/2022] Open
Abstract
Recent studies have suggested a protective role of physiological β-amyloid autoantibodies (Aβ-autoantibodies) in Alzheimer’s disease (AD). However, the determination of both free and dissociated Aβ-autoantibodies in serum hitherto has yielded inconsistent results regarding their function and possible biomarker value. Here we report the application of a new sandwich enzyme-linked immunosorbent assay (ELISA) for the determination of antigen-bound Aβ-autoantibodies (intact Aβ-IgG immune complexes) in serum and cerebrospinal fluid (CSF) of a total number of 112 AD patients and age- and gender-matched control subjects. Both serum and CSF levels of Aβ-IgG immune complexes were found to be significantly higher in AD patients compared to control subjects. Moreover, the levels of Aβ-IgG complexes were negatively correlated with the cognitive status across the groups, increasing with declining cognitive test performance of the subjects. Our results suggest a contribution of IgG-type autoantibodies to Aβ clearance in vivo and an increased immune response in AD, which may be associated with deficient Aβ-IgG removal. These findings may contribute to elucidating the role of Aβ-autoantibodies in AD pathophysiology and their potential application in AD diagnosis.
Collapse
Affiliation(s)
- Madalina Maftei
- Laboratory of Analytical Chemistry and Biopolymer Structure Analysis, Department of Chemistry, University of Konstanz, Konstanz, Germany
- Steinbeis Research Center for Biopolymer Analysis, University of Konstanz, Konstanz, Germany
| | - Franka Thurm
- Department of Psychology, University of Konstanz, Konstanz, Germany
- Clinical and Biological Psychology, Institute of Psychology and Education, University of Ulm, Ulm, Germany
- Department of Psychology, TU Dresden, Dresden, Germany
| | | | | | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Thomas Elbert
- Department of Psychology, University of Konstanz, Konstanz, Germany
| | - Iris-Tatjana Kolassa
- Clinical and Biological Psychology, Institute of Psychology and Education, University of Ulm, Ulm, Germany
- Zukunftskolleg, University of Konstanz, Konstanz, Germany
- * E-mail: (MM); (I-TK); (CAFVA); (MP)
| | - Michael Przybylski
- Laboratory of Analytical Chemistry and Biopolymer Structure Analysis, Department of Chemistry, University of Konstanz, Konstanz, Germany
- * E-mail: (MM); (I-TK); (CAFVA); (MP)
| | - Marilena Manea
- Laboratory of Analytical Chemistry and Biopolymer Structure Analysis, Department of Chemistry, University of Konstanz, Konstanz, Germany
- Zukunftskolleg, University of Konstanz, Konstanz, Germany
- * E-mail: (MM); (I-TK); (CAFVA); (MP)
| | | |
Collapse
|
49
|
Lu C, Guo Y, Yan J, Luo Z, Luo HB, Yan M, Huang L, Li X. Design, Synthesis, and Evaluation of Multitarget-Directed Resveratrol Derivatives for the Treatment of Alzheimer’s Disease. J Med Chem 2013; 56:5843-59. [DOI: 10.1021/jm400567s] [Citation(s) in RCA: 183] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Chuanjun Lu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yueyan Guo
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jun Yan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zonghua Luo
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Hai-Bin Luo
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Ming Yan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Ling Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Xingshu Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| |
Collapse
|
50
|
Piccinni A, Origlia N, Veltri A, Vizzaccaro C, Marazziti D, Vanelli F, Moroni I, Domenici L, Dell'Osso L. Neurodegeneration, β-amyloid and mood disorders: state of the art and future perspectives. Int J Geriatr Psychiatry 2013; 28:661-71. [PMID: 22996674 DOI: 10.1002/gps.3879] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 08/02/2012] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Depression may increase the risk of developing Alzheimer's disease (AD). Recent studies have shown modifications in blood beta-amyloid (Aβ) levels in depressed patients. This literature review examines the potential relationship between Aβ-mediated neurotoxicity and pathophysiology of mood disorders. DESIGN We conducted a review of the literature focusing on recent studies reporting alterations of plasma and serum Aβ peptides levels in patients suffering from mood disorders. RESULTS Different data suggest that patients with mood disorders are at great risk of developing cognitive impairment and dementia. In particular, low plasma levels of Aβ42 peptide and a high Aβ40/Aβ42 ratio have been found in depressed patients. In addition, changes in Aβ protein levels in patients with mood disorders have been associated with the severity of cognitive impairment and correlated positively with the number of episodes and severity of illness course. CONCLUSIONS Given the intriguing association between change in plasma level of Aβ, depression and cognitive impairment, future work should focus on the relationship between Aβ peripheral level(s), biomarkers of neurodegeneration and development of dementia in patients affected by mood disorders.
Collapse
Affiliation(s)
- Armando Piccinni
- Department of Psychiatry, Neurobiology, Pharmacology and Biotechnology, University of Pisa, Pisa, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|