151
|
Oberlin LE, Erickson KI, Mackey R, Klunk WE, Aizenstein H, Lopresti BJ, Kuller LH, Lopez OL, Snitz BE. Peripheral inflammatory biomarkers predict the deposition and progression of amyloid-β in cognitively unimpaired older adults. Brain Behav Immun 2021; 95:178-189. [PMID: 33737171 PMCID: PMC8647033 DOI: 10.1016/j.bbi.2021.03.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 02/23/2021] [Accepted: 03/10/2021] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION Systemic inflammation has been increasingly implicated in the pathogenesis of Alzheimer's disease (AD), yet the mechanistic and temporal specificity of this relationship is poorly understood. We aimed to characterize the cross-sectional and longitudinal associations between peripheral inflammatory biomarkers, cognition, and Aβ deposition in oldest-old cognitively unimpaired (CU) adults. METHODS A large sample of 139 CU older adults (mean age (range) = 85.4 (82-95)) underwent neuropsychological testing, Pittsburgh compound-B (PiB)-PET imaging and structural MRI. Hierarchical regression models examined associations between circulating inflammatory biomarkers (Interleukin-6 (IL-6), soluble Tumor Necrosis Factor receptors 1 and 2 (sTNFr1 and sTNFr2), soluble cluster of differentiation 14 (sCD14), C-reactive protein (CRP)), cognition, and global and regional Aβ deposition at baseline and over follow-up. Indices of preclinical disease, including pathologic Aβ status and hippocampal volume, were incorporated to assess conditional associations. RESULTS At baseline evaluation, higher concentrations of IL-6 and sTNFr2 were associated with greater global Aβ burden in those with lower hippocampal volume. In longitudinal models, IL-6 predicted subsequent conversion to MCI and both IL-6 and CRP predicted greater change in global and regional Aβ deposition specifically among participants PiB-positive at baseline. These relationships withstood adjustment for demographic factors, anti-hypertensive medication use, history of diabetes, heart disease, APOE ε4 carrier status, and white matter lesions. DISCUSSION In a large prospective sample of CU adults aged 80 and over, peripheral inflammatory biomarkers were associated with and predictive of the progression of Aβ deposition. This was specific to those with biomarker evidence of preclinical AD at baseline, supporting recent evidence of disease-state-dependent differences in inflammatory expression profiles. Chronic, low-level systemic inflammation may exacerbate the deposition of Aβ pathology among those with emerging disease processes, and place individuals at a higher risk of developing clinically significant cognitive impairment.
Collapse
Affiliation(s)
| | - Kirk I. Erickson
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA,Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA,College of Science, Health, Engineering and Education, Murdoch University, Perth, Australia
| | - Rachel Mackey
- Premier Applied Sciences, Premier Inc., Charlotte, North Carolina,Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA
| | - William E. Klunk
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA
| | | | | | - Lewis H. Kuller
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA
| | - Oscar L. Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA,Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - Beth E. Snitz
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
152
|
Hale JM, Bijlsma MJ, Lorenti A. Does postponing retirement affect cognitive function? A counterfactual experiment to disentangle life course risk factors. SSM Popul Health 2021; 15:100855. [PMID: 34258375 PMCID: PMC8255239 DOI: 10.1016/j.ssmph.2021.100855] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 05/31/2021] [Accepted: 06/24/2021] [Indexed: 11/26/2022] Open
Abstract
Evidence suggests that contemporaneous labor force participation affects cognitive function; however, it is unclear whether it is employment itself or endogenous factors related to individuals’ likelihood of employment that protects against cognitive decline. We exploit innovations in counterfactual causal inference to disentangle the effect of postponing retirement on later-life cognitive function from the effects of other life-course factors. With the U.S. Health and Retirement Study (1996–2014, n = 20,469), we use the parametric g-formula to estimate the effect of postponing retirement to age 67. We also study whether the benefit of postponing retirement is affected by gender, education, and/or occupation, and whether retirement affects cognitive function through depressive symptoms or comorbidities. We find that postponing retirement is protective against cognitive decline, accounting for other life-course factors (population: 0.34, 95% confidence interval (CI): 0.20,0.47; individual: 0.43, 95% CI: 0.26,0.60). The extent of the protective effect depends on subgroup, with the highest educated experiencing the greatest mitigation of cognitive decline (individual: 50%, 95% CI: 32%,71%). By using innovative models that better reflect the empirical reality of interconnected life-course processes, this work makes progress in understanding how retirement affects cognitive function. Research is inconclusive as to employments' protection against cognitive decline. Causal inference models can better reflect interconnected life-course processes. The parametric g-formula shows a substantial protective effect of ongoing employment. Protective effect holds for all subgroups, but is greatest for the highest educated.
Collapse
Affiliation(s)
- Jo Mhairi Hale
- School of Geography and Sustainable Development, University of St Andrews, Scotland, UK.,Max Planck Institute for Demographic Research, Rostock, Germany
| | - Maarten J Bijlsma
- Max Planck Institute for Demographic Research, Rostock, Germany.,Groningen Research Institute of Pharmacy, Unit Pharmacotherapy, Epidemiology & Economics (PTEE), University of Groningen, The Netherlands
| | - Angelo Lorenti
- Max Planck Institute for Demographic Research, Rostock, Germany
| |
Collapse
|
153
|
Detecting retinal cell stress and apoptosis with DARC: Progression from lab to clinic. Prog Retin Eye Res 2021; 86:100976. [PMID: 34102318 DOI: 10.1016/j.preteyeres.2021.100976] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 12/15/2022]
Abstract
DARC (Detection of Apoptosing Retinal Cells) is a retinal imaging technology that has been developed within the last 2 decades from basic laboratory science to Phase 2 clinical trials. It uses ANX776 (fluorescently labelled Annexin A5) to identify stressed and apoptotic cells in the living eye. During its development, DARC has undergone biochemistry optimisation, scale-up and GMP manufacture and extensive preclinical evaluation. Initially tested in preclinical glaucoma and optic neuropathy models, it has also been investigated in Alzheimer, Parkinson's and Diabetic models, and used to assess efficacy of therapies. Progression to clinical trials has not been speedy. Intravenous ANX776 has to date been found to be safe and well-tolerated in 129 patients, including 16 from Phase 1 and 113 from Phase 2. Results on glaucoma and AMD patients have been recently published, and suggest DARC with an AI-aided algorithm can be used to predict disease activity. New analyses of DARC in GA prediction are reported here. Although further studies are needed to validate these findings, it appears there is potential of the technology to be used as a biomarker. Much larger clinical studies will be needed before it can be considered as a diagnostic, although the relatively non-invasive nature of the nasal as opposed to intravenous administration would widen its acceptability in the future as a screening tool. This review describes DARC development and its progression into Phase 2 clinical trials from lab-based research. It discusses hypotheses, potential challenges, and regulatory hurdles in translating technology.
Collapse
|
154
|
Comparative Performance and Neuropathologic Validation of the AD8 Dementia Screening Instrument. Alzheimer Dis Assoc Disord 2021; 34:112-117. [PMID: 31725472 DOI: 10.1097/wad.0000000000000362] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND/OBJECTIVE The AD8 informant-based screening instrument has been validated with molecular biomarkers of Alzheimer disease (AD) but not with the gold standard of neuropathologic AD. The objective of this study was to validate the AD8 with neuropathologic AD and compare its predictive performance with that of the Mini-Mental State Examination and both participant-derived and informant-derived subjective memory complaint (SMC) regarding the participant. METHODS This longitudinal cohort study at the Knight Alzheimer Disease Research Center at Washington University included 230 participants, ages 50 to 91 years, who later had a neuropathologic examination. Four dementia screening instruments from their baseline assessment were evaluated: the AD8, Mini-Mental State Examination, participant SMC, and informant SMC. The primary outcome was a neuropathologic diagnosis of AD. RESULTS The average participant age at baseline was 80.4 years, 48% were female. All 4 dementia screening tests were predictive of neuropathologic AD. There was no significant difference in the predictive performance of the AD8 compared with the other instruments, but the AD8 had superior sensitivity and combined positive and negative predictive values. CONCLUSION The AD8 is a brief and sensitive screening instrument that may facilitate earlier and more accurate AD diagnosis in a variety of care settings.
Collapse
|
155
|
Pinz MP, Vogt AG, da Costa Rodrigues K, Dos Reis AS, Duarte LFB, Fronza MG, Domingues WB, Blodorn EB, Alves D, Campos VF, Savegnago L, Wilhelm EA, Luchese C. Effect of a purine derivative containing selenium to improve memory decline and anxiety through modulation of the cholinergic system and Na +/K +-ATPase in an Alzheimer's disease model. Metab Brain Dis 2021; 36:871-888. [PMID: 33651275 DOI: 10.1007/s11011-021-00703-w] [Citation(s) in RCA: 12] [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: 09/20/2020] [Accepted: 02/22/2021] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is a worldwide problem, and there are currently no treatments that can stop this disease. To investigate the binding affinity of 6-((4-fluorophenyl) selanyl)-9H-purine (FSP) with acetylcholinesterase (AChE), to verify the effects of FSP in an AD model in mice and to evaluate the toxicological potential of this compound in mice. The binding affinity of FSP with AChE was investigated by molecular docking analyses. The AD model was induced by streptozotocin (STZ) in Swiss mice after FSP treatment (1 mg/kg, intragastrically (i.g.)), 1st-10th day of the experimental protocol. Anxiety was evaluated in an elevated plus maze test, and memory impairment was evaluated in the Y-maze, object recognition and step-down inhibitory avoidance tasks. The cholinergic system was investigated based on by looking at expression and activity of AChE and expression of choline acetyltransferase (ChAT). We evaluated expression and activity of Na+/K+-ATPase. For toxicological analysis, animals received FSP (300 mg/kg, i.g.) and aspartate aminotransferase, alanine aminotransferase activities were determined in plasma and δ-aminolevulinate dehydratase activity in brain and liver. FSP interacts with residues of the AChE active site. FSP mitigated the induction of anxiety and memory impairment caused by STZ. FSP protected cholinergic system dysfunction and reduction of activity and expression of Na+/K+-ATPase. FSP did not modify toxicological parameters evaluated and did not cause the death of mice. FSP protected against anxiety, learning and memory impairment with involvement of the cholinergic system and Na+/K+-ATPase in these actions.
Collapse
Affiliation(s)
- Mikaela Peglow Pinz
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP96010-900, Brazil
| | - Ane Gabriela Vogt
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP96010-900, Brazil
| | - Karline da Costa Rodrigues
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP96010-900, Brazil
| | - Angélica Schiavom Dos Reis
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP96010-900, Brazil
| | - Luis Fernando Barbosa Duarte
- Programa de Pós-Graduação em Química, Laboratório de Síntese Orgânica Limpa (LASOL), CCQFA, UFPel, Pelotas, RS, CEP 96010-900, Brazil
| | - Mariana Gallio Fronza
- Programa de Pós-Graduação em Biotecnologia, GPN, CDTec, UFPel, Pelotas, RS, CEP 96010-900, Brazil
| | - William Borges Domingues
- Programa de Pós-Graduação em Biotecnologia, Laboratório de Genômica Estrutural, Centro de Desenvolvimento Tecnológico (CDTec), UFPel, Pelotas, RS, CEP 96010-900, Brazil
| | - Eduardo Bierhaus Blodorn
- Programa de Pós-Graduação em Biotecnologia, Laboratório de Genômica Estrutural, Centro de Desenvolvimento Tecnológico (CDTec), UFPel, Pelotas, RS, CEP 96010-900, Brazil
| | - Diego Alves
- Programa de Pós-Graduação em Química, Laboratório de Síntese Orgânica Limpa (LASOL), CCQFA, UFPel, Pelotas, RS, CEP 96010-900, Brazil
| | - Vinicius Farias Campos
- Programa de Pós-Graduação em Biotecnologia, Laboratório de Genômica Estrutural, Centro de Desenvolvimento Tecnológico (CDTec), UFPel, Pelotas, RS, CEP 96010-900, Brazil
| | - Lucielli Savegnago
- Programa de Pós-Graduação em Biotecnologia, GPN, CDTec, UFPel, Pelotas, RS, CEP 96010-900, Brazil
| | - Ethel Antunes Wilhelm
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP96010-900, Brazil.
| | - Cristiane Luchese
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA), Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP96010-900, Brazil.
| |
Collapse
|
156
|
Rajan KB, Weuve J, Barnes LL, McAninch EA, Wilson RS, Evans DA. Population estimate of people with clinical Alzheimer's disease and mild cognitive impairment in the United States (2020-2060). Alzheimers Dement 2021; 17:1966-1975. [PMID: 34043283 DOI: 10.1002/alz.12362] [Citation(s) in RCA: 639] [Impact Index Per Article: 159.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 03/29/2021] [Accepted: 04/05/2021] [Indexed: 02/04/2023]
Abstract
INTRODUCTION The estimate of people with clinical Alzheimer's disease (AD) and mild cognitive impairment provides an understanding of the disease burden. METHODS We estimated people with cognitive impairment using a quasibinomial regression model in 10,342 participants with cognitive test scores. RESULTS The 2020 US Census-adjusted prevalence of clinical AD was 11.3% (95% confidence interval [CI] = 10.7-11.9): 10.0% among non-Hispanic Whites, 14.0% among Hispanics, and 18.6% among non-Hispanic Blacks. We estimate that in 2020, 6.07 (95% CI = 5.75-6.38) million people were living with clinical AD, which increases to 13.85 (95% CI = 12.98-14.74) million in 2060, 423% higher among Hispanics, 192% higher among Blacks, and 63% higher among Whites. However, there are predicted to be more significant increases in later years among those over 85 and women compared to men. DISCUSSION The number of people with clinical AD will increase as the "baby boom" generation reaches older ages, exerting a strong upward influence on disease burden.
Collapse
Affiliation(s)
- Kumar B Rajan
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, Illinois, USA
| | - Jennifer Weuve
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Lisa L Barnes
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA
| | - Elizabeth A McAninch
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, Illinois, USA
| | - Robert S Wilson
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois, USA
| | - Denis A Evans
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
157
|
Amini Y, Saif N, Greer C, Hristov H, Isaacson R. The Role of Nutrition in Individualized Alzheimer's Risk Reduction. Curr Nutr Rep 2021; 9:55-63. [PMID: 32277428 DOI: 10.1007/s13668-020-00311-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Decades of research suggests nutritional interventions can be an effective tool for reducing risk of Alzheimer's disease (AD), especially as part of an individualized clinical management plan. This review aims to emphasize new findings examining how specific dietary changes may delay or possibly prevent AD onset, and highlight how interventions can be adopted in clinical practice based on emerging principles of precision medicine. RECENT FINDINGS Specific dietary patterns and varied nutrient combinations can have a protective effect on brain health, promote cognitive function, and mediate the comorbidity of chronic conditions associated with increased AD risk. Individuals at risk for AD may see a greater impact of evidence-based dietary changes when initiated earlier in the AD spectrum. Depending on individual clinical profiles, incorporation of nutrition strategies is an essential component of an AD risk reduction plan in clinical practice.
Collapse
Affiliation(s)
- Yasmin Amini
- Department of Neurology, Weill Cornell Medicine & New York-Presbyterian, 428 East 72th St, Suite 500, New York, NY, 10021, USA
- Department of Sociomedical Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Nabeel Saif
- Department of Neurology, Weill Cornell Medicine & New York-Presbyterian, 428 East 72th St, Suite 500, New York, NY, 10021, USA
| | - Christine Greer
- Department of Neurology, Weill Cornell Medicine & New York-Presbyterian, 428 East 72th St, Suite 500, New York, NY, 10021, USA
| | - Hollie Hristov
- Department of Neurology, Weill Cornell Medicine & New York-Presbyterian, 428 East 72th St, Suite 500, New York, NY, 10021, USA
| | - Richard Isaacson
- Department of Neurology, Weill Cornell Medicine & New York-Presbyterian, 428 East 72th St, Suite 500, New York, NY, 10021, USA.
| |
Collapse
|
158
|
Alsharif AA, Wei L, Ma T, Man KKC, Lau WCY, Brauer R, Almetwazi M, Howard R, Wong ICK. Prevalence and Incidence of Dementia in People with Diabetes Mellitus. J Alzheimers Dis 2021; 75:607-615. [PMID: 32310163 DOI: 10.3233/jad-191115] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Few studies have shown that an increased risk of dementia is associated with diabetes mellitus. OBJECTIVE To estimate the prevalence and incidence of dementia in people with diabetes in primary care in the UK. METHODS We conducted a descriptive study using the UK The Health Improvement Network (THIN) database. People diagnosed with diabetes from 2000 to 2016 were included in the study. Prevalence and incidence rates of dementia were calculated annually, stratified by age and gender. RESULTS The prevalence of dementia was 0.424% [95% CI (0.420%-0.427%)] in 2000 and 2.508% [95% CI (2.501%-2.515%)] in 2016. The highest prevalence was in those aged 85+ from 2.9% [95% CI (2.890%-2.974%)] in 2000 to 11.3% [95% CI (11.285%-11.384%)] in 2016. The incidence of dementia increased 3.7 times, from 0.181 cases per 100 persons [95% CI (0.179-0.183)] in 2000 to 0.683 cases per 100 persons [95% CI (0.679-0.686)] in 2016, respectively. Women had a higher prevalence and incidence of dementia than men 3.138% [95% CI (3.127%-3.150%)] versus 2.014% [95% CI (2.006%-2.022%)] and 0.820 [95% CI (0.814-0.826)] versus 0.576 cases per 100 persons [95% CI (0.571-0.580)] in 2016, respectively. CONCLUSION There was a trend of increasing prevalence and incidence of dementia in people with diabetes over the period of 2000 to 2016. This study adds to the evidence on dementia prevalence and incidence, particularly in the diabetic population.
Collapse
Affiliation(s)
- Alaa A Alsharif
- Department of Pharmacy Practice, Faculty of Pharmacy, Princess Norah Bint Abdulrahman University, Riyadh, Kingdom of Saudi Arabia.,Research Department of Practice and Policy, University College London School of Pharmacy, London, United Kingdom
| | - Li Wei
- Research Department of Practice and Policy, University College London School of Pharmacy, London, United Kingdom
| | - Tiantian Ma
- Research Department of Practice and Policy, University College London School of Pharmacy, London, United Kingdom
| | - Kenneth K C Man
- Research Department of Practice and Policy, University College London School of Pharmacy, London, United Kingdom.,Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.,Department of Medical Informatics, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Wallis C Y Lau
- Research Department of Practice and Policy, University College London School of Pharmacy, London, United Kingdom.,Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Ruth Brauer
- Research Department of Practice and Policy, University College London School of Pharmacy, London, United Kingdom
| | - Mansour Almetwazi
- Clinical Pharmacy Department, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Rob Howard
- Division of Psychiatry, University College London, London, United Kingdom
| | - Ian C K Wong
- Research Department of Practice and Policy, University College London School of Pharmacy, London, United Kingdom.,Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| |
Collapse
|
159
|
Da Mesquita S, Herz J, Wall M, Dykstra T, de Lima KA, Norris GT, Dabhi N, Kennedy T, Baker W, Kipnis J. Aging-associated deficit in CCR7 is linked to worsened glymphatic function, cognition, neuroinflammation, and β-amyloid pathology. SCIENCE ADVANCES 2021; 7:eabe4601. [PMID: 34020948 PMCID: PMC8139596 DOI: 10.1126/sciadv.abe4601] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 04/02/2021] [Indexed: 05/02/2023]
Abstract
Aging leads to a progressive deterioration of meningeal lymphatics and peripheral immunity, which may accelerate cognitive decline. We hypothesized that an age-related reduction in C-C chemokine receptor type 7 (CCR7)-dependent egress of immune cells through the lymphatic vasculature mediates some aspects of brain aging and potentially exacerbates cognitive decline and Alzheimer's disease-like brain β-amyloid (Aβ) pathology. We report a reduction in CCR7 expression by meningeal T cells in old mice that is linked to increased effector and regulatory T cells. Hematopoietic CCR7 deficiency mimicked the aging-associated changes in meningeal T cells and led to reduced glymphatic influx and cognitive impairment. Deletion of CCR7 in 5xFAD transgenic mice resulted in deleterious neurovascular and microglial activation, along with increased Aβ deposition in the brain. Treating old mice with anti-CD25 antibodies alleviated the exacerbated meningeal regulatory T cell response and improved cognitive function, highlighting the therapeutic potential of modulating meningeal immunity to fine-tune brain function in aging and in neurodegenerative diseases.
Collapse
Affiliation(s)
- Sandro Da Mesquita
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA.
| | - Jasmin Herz
- Center for Brain Immunology and Glia (BIG), Washington University in St. Louis, St. Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Morgan Wall
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
| | - Taitea Dykstra
- Center for Brain Immunology and Glia (BIG), Washington University in St. Louis, St. Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Kalil Alves de Lima
- Center for Brain Immunology and Glia (BIG), Washington University in St. Louis, St. Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Geoffrey T Norris
- Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Nisha Dabhi
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
| | - Tatiana Kennedy
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
| | - Wendy Baker
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA
| | - Jonathan Kipnis
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA.
- Center for Brain Immunology and Glia (BIG), Washington University in St. Louis, St. Louis, MO, USA
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
160
|
Chen L, Wei Z, Chan KWY, Li Y, Suchal K, Bi S, Huang J, Xu X, Wong PC, Lu H, van Zijl PCM, Li T, Xu J. D-Glucose uptake and clearance in the tauopathy Alzheimer's disease mouse brain detected by on-resonance variable delay multiple pulse MRI. J Cereb Blood Flow Metab 2021; 41:1013-1025. [PMID: 32669023 PMCID: PMC8054725 DOI: 10.1177/0271678x20941264] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/29/2020] [Accepted: 06/17/2020] [Indexed: 12/11/2022]
Abstract
In this study, we applied on-resonance variable delay multiple pulse (onVDMP) MRI to study D-glucose uptake in a mouse model of Alzheimer's disease (AD) tauopathy and demonstrated its feasibility in discriminating AD mice from wild-type mice. The D-glucose uptake in the cortex of AD mice (1.70 ± 1.33%) was significantly reduced compared to that of wild-type mice (5.42 ± 0.70%, p = 0.0051). Also, a slower D-glucose uptake rate was found in the cerebrospinal fluid (CSF) of AD mice (0.08 ± 0.01 min-1) compared to their wild-type counterpart (0.56 ± 0.1 min-1, p < 0.001), which suggests the presence of an impaired glucose transporter on both blood-brain and blood-CSF barriers of these AD mice. Clearance of D-glucose was observed in the CSF of wild-type mice but not AD mice, which suggests dysfunction of the glymphatic system in the AD mice. The results in this study indicate that onVDMP MRI could be a cost-effective and widely available method for simultaneously evaluating glucose transporter and glymphatic function of AD. This study also suggests that tau protein affects the D-glucose uptake and glymphatic impairment in AD at a time point preceding neurofibrillary tangle pathology.
Collapse
Affiliation(s)
- Lin Chen
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhiliang Wei
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kannie WY Chan
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Yuguo Li
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kapil Suchal
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sheng Bi
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jianpan Huang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Xiang Xu
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Philip C Wong
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hanzhang Lu
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter CM van Zijl
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tong Li
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jiadi Xu
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD, USA
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
161
|
Petkus AJ, Gomez ME. The importance of social support, engagement in leisure activities, and cognitive reserve in older adulthood. Int Psychogeriatr 2021; 33:433-435. [PMID: 34057062 DOI: 10.1017/s1041610220003336] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Andrew J Petkus
- Department of Neurology, University of Southern California, Los Angeles, CA, USA
| | - Megan E Gomez
- Psychology Service, VA Long Beach Healthcare System, Long Beach, CA, USA
| |
Collapse
|
162
|
Mielke MM. Consideration of Sex Differences in the Measurement and Interpretation of Alzheimer Disease-Related Biofluid-Based Biomarkers. J Appl Lab Med 2021; 5:158-169. [PMID: 31811073 DOI: 10.1373/jalm.2019.030023] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/23/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND The development of cerebrospinal fluid and blood-based biomarkers for Alzheimer disease (AD) and related disorders is rapidly progressing. Such biomarkers may be used clinically to screen the population, to enhance diagnosis, or to help determine prognosis. Although the use of precision medicine methods has contributed to enhanced understanding of the AD pathophysiological changes and development of assays, one aspect not commonly considered is sex differences. CONTENT There are several ways in which sex can affect the concentration or interpretation of biofluid biomarkers. For some markers, concentrations will vary by sex. For others, the concentrations might not vary by sex, but the impact or interpretation may vary by sex depending on the context of use (e.g., diagnostic vs prognostic). Finally, for others, there will be no sex differences in concentrations or their interpretation. This review will first provide a basis for sex differences, including differences in brain structure and function, and the means by which these differences could contribute to sex differences in biomarker concentrations. Next, the current state of sex differences in AD-related biofluid markers (i.e., amyloid-β, phosphorylated τ, total τ, neurofilament light chain, and neurogranin) will be reviewed. Lastly, factors that can lead to the misinterpretation of observed sex differences in biomarkers (either providing evidence for or against) will be considered. SUMMARY This review is intended to provide an impetus to consider sex differences in the measurement and interpretation of AD-related biofluid-based biomarkers.
Collapse
Affiliation(s)
- Michelle M Mielke
- Departments of Health Sciences Research and Neurology, Mayo Clinic, Rochester, MN
| |
Collapse
|
163
|
Sun N, Meng X, Liu Y, Song D, Jiang C, Cai J. Applications of brain organoids in neurodevelopment and neurological diseases. J Biomed Sci 2021; 28:30. [PMID: 33888112 PMCID: PMC8063318 DOI: 10.1186/s12929-021-00728-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 04/18/2021] [Indexed: 12/20/2022] Open
Abstract
A brain organoid is a self-organizing three-dimensional tissue derived from human embryonic stem cells or pluripotent stem cells and is able to simulate the architecture and functionality of the human brain. Brain organoid generation methods are abundant and continue to improve, and now, an in vivo vascularized brain organoid has been encouragingly reported. The combination of brain organoids with immune-staining and single-cell sequencing technology facilitates our understanding of brain organoids, including the structural organization and the diversity of cell types. Recent publications have reported that brain organoids can mimic the dynamic spatiotemporal process of early brain development, model various human brain disorders, and serve as an effective preclinical platform to test and guide personalized treatment. In this review, we introduce the current state of brain organoid differentiation strategies, summarize current progress and applications in the medical domain, and discuss the challenges and prospects of this promising technology.
Collapse
Affiliation(s)
- Nan Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Xiangqi Meng
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Yuxiang Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Dan Song
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Chuanlu Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China.
| | - Jinquan Cai
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China.
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, 171 65, Stockholm, Sweden.
| |
Collapse
|
164
|
Rabipour S, Rajagopal S, Pasvanis S, Rajah MN. Generalization of memory-related brain function in asymptomatic older women with a family history of late onset Alzheimer's Disease: Results from the PREVENT-AD Cohort. Neurobiol Aging 2021; 104:42-56. [PMID: 33964608 DOI: 10.1016/j.neurobiolaging.2021.03.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/04/2021] [Accepted: 03/18/2021] [Indexed: 11/18/2022]
Abstract
Late-onset Alzheimer's disease (AD) disproportionately affects women compared to men. Episodic memory decline is one of the earliest and most pronounced deficits observed in AD. However, it remains unclear whether sex influences episodic memory-related brain function in cognitively intact older adults at risk of developing AD. Here we used task-based multivariate partial least squares analysis to examine sex differences in episodic memory-related brain activity and brain activity-behavior correlations in a matched sample of cognitively intact older women and men with a family history of AD from the PREVENT-AD cohort study in Montreal, Canada (Mage=63.03±3.78; Meducation=15.41±3.40). We observed sex differences in task-related brain activity and brain activity-behavior correlations during the encoding of object-location associative memories and object-only item memory, and the retrieval of object only item memories. Our findings suggest a generalization of episodic memory-related brain activation and performance in women compared to men. Follow up analyses should test for sex differences in the relationship between brain activity patterns and performance longitudinally, in association with risk factors for AD development. This article is part of the Virtual Special Issue titled COGNITIVE NEUROSCIENCE OF HEALTHY AND PATHOLOGICAL AGING. The full issue can be found on ScienceDirect at https://www.sciencedirect.com/journal/neurobiology-of-aging/special-issue/105379XPWJP.
Collapse
Affiliation(s)
- Sheida Rabipour
- Centre for Cerebral Imaging, Douglas Hospital Research Centre, Montreal, Canada; Department of Psychiatry, McGill University, Montreal, Canada
| | | | - Stamatoula Pasvanis
- Centre for Cerebral Imaging, Douglas Hospital Research Centre, Montreal, Canada
| | - M Natasha Rajah
- Centre for Cerebral Imaging, Douglas Hospital Research Centre, Montreal, Canada; Department of Psychiatry, McGill University, Montreal, Canada.
| |
Collapse
|
165
|
Abstract
This article describes the public health impact of Alzheimer's disease (AD), including incidence and prevalence, mortality and morbidity, use and costs of care, and the overall impact on caregivers and society. The Special Report discusses the challenges of providing equitable health care for people with dementia in the United States. An estimated 6.2 million Americans age 65 and older are living with Alzheimer's dementia today. This number could grow to 13.8 million by 2060 barring the development of medical breakthroughs to prevent, slow or cure AD. Official death certificates recorded 121,499 deaths from AD in 2019, the latest year for which data are available, making Alzheimer's the sixth-leading cause of death in the United States and the fifth-leading cause of death among Americans age 65 and older. Between 2000 and 2019, deaths from stroke, heart disease and HIV decreased, whereas reported deaths from AD increased more than 145%. This trajectory of deaths from AD was likely exacerbated in 2020 by the COVID-19 pandemic. More than 11 million family members and other unpaid caregivers provided an estimated 15.3 billion hours of care to people with Alzheimer's or other dementias in 2020. These figures reflect a decline in the number of caregivers compared with a decade earlier, as well as an increase in the amount of care provided by each remaining caregiver. Unpaid dementia caregiving was valued at $256.7 billion in 2020. Its costs, however, extend to family caregivers' increased risk for emotional distress and negative mental and physical health outcomes - costs that have been aggravated by COVID-19. Average per-person Medicare payments for services to beneficiaries age 65 and older with AD or other dementias are more than three times as great as payments for beneficiaries without these conditions, and Medicaid payments are more than 23 times as great. Total payments in 2021 for health care, long-term care and hospice services for people age 65 and older with dementia are estimated to be $355 billion. Despite years of efforts to make health care more equitable in the United States, racial and ethnic disparities remain - both in terms of health disparities, which involve differences in the burden of illness, and health care disparities, which involve differences in the ability to use health care services. Blacks, Hispanics, Asian Americans and Native Americans continue to have a higher burden of illness and lower access to health care compared with Whites. Such disparities, which have become more apparent during COVID-19, extend to dementia care. Surveys commissioned by the Alzheimer's Association recently shed new light on the role of discrimination in dementia care, the varying levels of trust between racial and ethnic groups in medical research, and the differences between groups in their levels of concern about and awareness of Alzheimer's disease. These findings emphasize the need to increase racial and ethnic diversity in both the dementia care workforce and in Alzheimer's clinical trials.
Collapse
|
166
|
Hamza SA, Asif S, Bokhari SAH. Oral health of individuals with dementia and Alzheimer's disease: A review. J Indian Soc Periodontol 2021; 25:96-101. [PMID: 33888939 PMCID: PMC8041071 DOI: 10.4103/jisp.jisp_287_20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 11/30/2020] [Accepted: 12/12/2020] [Indexed: 11/04/2022] Open
Abstract
This paper explores the epidemiological evidence about oral health of individuals with neurodegenerative conditions of Alzheimer's disease (AD) and dementia. PubMed, Web of Science, and Scopus were searched to identify the relevant research papers published during January 2012 to June 2020. All cross-sectional, case-control, and cohort studies reporting oral and dental morbid conditions for status and association with AD and dementia were explored. The explored literature from 22 studies shows that oral health parameters of oral health and levels of oral inflammatory markers were deranged and exaggerated in patients suffering from AD and dementia. Many studies have observed poor oral hygiene as result of lack or irregularity in toothbrushing. Regarding decayed, missing, and filled teeth status in AD/dementia populations, no significant difference is reported. Periodontal diseases have been noted at raised levels in AD and dementia patients and shown progression with aggravation in neurological disorders. Both edentulousness and low chewing efficacies are associated with low cognition. Stomatitis and coated tongue and other oral pathologies are significantly higher in AD patients. AD patients have demonstrated higher bacterial load and inflammation levels than controls, and consequently, inflammatory biomarker levels are also raised. AD patients have reduced salivary secretions and with low buffering capacity. Evidence from the current literature update postulates that individuals suffering from AD and dementia have special oral health-care needs. Appropriate oral health management may thus significantly improve their oral health-related and general quality of life.
Collapse
Affiliation(s)
- Syed Ameer Hamza
- Department of Oral Medicine, University Medical and Dental College, The University of Faisalabad, Faisalabad, Pakistan
| | - Saba Asif
- Department of Periodontology, College of Dentistry, Sharif Medical and Dental College Lahore, Pakistan
| | - Syed Akhtar Hussain Bokhari
- Department of Preventive Dental Sciences, Committee for Postgraduate Studies and Research College of Dentistry, King Faisal University Al-Ahsa, Saudi Arabia
| |
Collapse
|
167
|
Forecasting of the Prevalence of Dementia Using the LSTM Neural Network in Taiwan. MATHEMATICS 2021. [DOI: 10.3390/math9050488] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The World Health Organization has urged countries to prioritize dementia in their public health policies. Dementia poses a tremendous socioeconomic burden, and the accurate prediction of the annual increase in prevalence is essential for establishing strategies to cope with its effects. The present study established a model based on the architecture of the long short-term memory (LSTM) neural network for predicting the number of dementia cases in Taiwan, which considers the effects of age and sex on the prevalence of dementia. The LSTM network is a variant of recurrent neural networks (RNNs), which possesses a special gate structure and avoids the problems in RNNs of gradient explosion, gradient vanishing, and long-term memory failure. A number of patients diagnosed as having dementia from 1997 to 2017 was collected in annual units from a data set extracted from the Health Insurance Database of the Ministry of Health and Welfare in Taiwan. To further verify the validity of the proposed model, the LSTM network was compared with three types of models: statistical models (exponential smoothing (ETS), autoregressive integrated moving average model (ARIMA), trigonometric seasonality, Box–Cox transformation, autoregressive moving average errors, and trend seasonal components model (TBATS)), hybrid models (support vector regression (SVR), particle swarm optimization–based support vector regression (PSOSVR)), and deep learning model (artificial neural networks (ANN)). The mean absolute percentage error (MAPE), root-mean-square error (RMSE), mean absolute error (MAE), and R-squared (R2) were used to evaluate the model performances. The results indicated that the LSTM network has higher prediction accuracy than the three types of models for forecasting the prevalence of dementia in Taiwan.
Collapse
|
168
|
Ashford MT, Veitch DP, Neuhaus J, Nosheny RL, Tosun D, Weiner MW. The search for a convenient procedure to detect one of the earliest signs of Alzheimer's disease: A systematic review of the prediction of brain amyloid status. Alzheimers Dement 2021; 17:866-887. [PMID: 33583100 DOI: 10.1002/alz.12253] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/10/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Convenient, cost-effective tests for amyloid beta (Aβ) are needed to identify those at higher risk for developing Alzheimer's disease (AD). This systematic review evaluates recent models that predict dichotomous Aβ. (PROSPERO: CRD42020144734). METHODS We searched Embase and identified 73 studies from 29,581 for review. We assessed study quality using established tools, extracted information, and reported results narratively. RESULTS We identified few high-quality studies due to concerns about Aβ determination and analytical issues. The most promising convenient, inexpensive classifiers consist of age, apolipoprotein E genotype, cognitive measures, and/or plasma Aβ. Plasma Aβ may be sufficient if pre-analytical variables are standardized and scalable assays developed. Some models lowered costs associated with clinical trial recruitment or clinical screening. DISCUSSION Conclusions about models are difficult due to study heterogeneity and quality. Promising prediction models used demographic, cognitive/neuropsychological, imaging, and plasma Aβ measures. Further studies using standardized Aβ determination, and improved model validation are required.
Collapse
Affiliation(s)
- Miriam T Ashford
- Department of Veterans Affairs Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA.,Department of Veterans Affairs Medical Center, Center for Imaging and Neurodegenerative Diseases, San Francisco, California, USA
| | - Dallas P Veitch
- Department of Veterans Affairs Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA
| | - John Neuhaus
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| | - Rachel L Nosheny
- Department of Veterans Affairs Medical Center, Center for Imaging and Neurodegenerative Diseases, San Francisco, California, USA.,Department of Psychiatry, University of California San Francisco, San Francisco, California, USA
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Michael W Weiner
- Department of Veterans Affairs Medical Center, Northern California Institute for Research and Education, San Francisco, California, USA.,Department of Veterans Affairs Medical Center, Center for Imaging and Neurodegenerative Diseases, San Francisco, California, USA.,Department of Psychiatry, University of California San Francisco, San Francisco, California, USA.,Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA.,Department of Medicine, University of California San Francisco, San Francisco, California, USA.,Department of Neurology, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
169
|
Residue Interaction Network Analysis Predicts a Val24-Ile31 Interaction May be Involved in Preventing Amyloid-Beta (1-42) Primary Nucleation. Protein J 2021; 40:175-183. [PMID: 33566321 DOI: 10.1007/s10930-021-09965-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2021] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) patients could benefit from a more effective treatment than the current FDA-approved options. Because amyloid-beta (Aβ) is thought to play a central role in AD pathogenesis, many experimental drugs attempt to reduce Aβ-induced pathology. Preventing amyloid accumulation may be a more effective strategy than clearing Aβ plaques after they form. If preventing Aβ accumulation can treat or prevent AD, then understanding Aβ primary nucleation may aid rational drug design. This study examines Aβ residue interaction networks and reports network and structural observations that may provide insight into primary nucleation. While many studies identify structural features of Aβ that promote aggregation, this study reports features that may resist primary nucleation by examining Aβ42 studies in more and less polar solvents. In Aβ42 in a less polar solvent (PDB ID: 1IYT), Val24 and Ile31 have higher betweenness and residue centrality values. This may be due to a predicted interaction between Val24 and Ile31. Residues in the central hydrophobic cluster (CHC) of Aβ40 and Aβ42 had significantly higher betweenness values compared to the average betweenness of the structures, highlighting the CHC's reported role in oligomerization. The predicted interaction between Val24 and Ile31 may reduce the likelihood of primary nucleation of Aβ.
Collapse
|
170
|
Bacterial sepsis increases hippocampal fibrillar amyloid plaque load and neuroinflammation in a mouse model of Alzheimer's disease. Neurobiol Dis 2021; 152:105292. [PMID: 33556539 DOI: 10.1016/j.nbd.2021.105292] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/06/2020] [Accepted: 02/03/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Sepsis, a leading cause for intensive care unit admissions, causes both an acute encephalopathy and chronic brain dysfunction in survivors. A history of sepsis is also a risk factor for future development of dementia symptoms. Similar neuropathologic changes are associated with the cognitive decline of sepsis and Alzheimer's disease (AD), including neuroinflammation, neuronal death, and synaptic loss. Amyloid plaque pathology is the earliest pathological hallmark of AD, appearing 10 to 20 years prior to cognitive decline, and is present in 30% of people over 65. As sepsis is also more common in older adults, we hypothesized that sepsis might exacerbate amyloid plaque deposition and plaque-related injury, promoting the progression of AD-related pathology. METHODS We evaluated whether the brain's response to sepsis modulates AD-related neurodegenerative changes by driving amyloid deposition and neuroinflammation in mice. We induced polymicrobial sepsis by cecal ligation and puncture (CLP) in APP/PS1-21 mice, a model of AD-related β-amyloidosis. We performed CLP or sham surgery at plaque onset (2 months of age) and examined pathology 2 months after CLP in surviving mice. RESULTS Sepsis significantly increased fibrillar amyloid plaque formation in the hippocampus of APP/PS1-21 mice. Sepsis enhanced plaque-related astrocyte activation and complement C4b gene expression in the brain, both of which may play a role in modulating amyloid formation. CLP also caused large scale changes in the gut microbiome of APP/PS1 mice, which have been associated with a pro-amyloidogenic and neuroinflammatory state. CONCLUSIONS Our results suggest that experimental sepsis can exacerbate amyloid plaque deposition and plaque-related inflammation, providing a potential mechanism for increased dementia in older sepsis survivors.
Collapse
|
171
|
Yin P, Xue Y, Wang T, Zhong D, Li G. The Therapeutic Targets of Fingolimod (FTY720) Are Involved in Pathological Processes in the Frontal Cortex of Alzheimer's Disease Patients: A Network Pharmacology Study. Front Aging Neurosci 2021; 13:609679. [PMID: 33603656 PMCID: PMC7884771 DOI: 10.3389/fnagi.2021.609679] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/05/2021] [Indexed: 12/16/2022] Open
Abstract
Background: The sphingosine-1-phosphate receptor (S1PR) modulator fingolimod (FTY720), which is commonly used as an immunomodulator in multiple sclerosis treatment, has recently been found to reduce pathological changes in the brain tissue of Alzheimer's disease (AD) animal models, but this has yet to be verified in human brain tissue. In this study, network pharmacology methods were applied to determine the potential pharmacological mechanisms of fingolimod in the frontal cortex of AD patients. Methods: The pharmacological macromolecular targets of fingolimod and fingolimod phosphate were downloaded from SwissTarget and DrugBank. Systematic intersection analysis of the expression profiles of brain frontal cortex tissues (423 AD tissues and 266 control tissues) was performed to obtain AD-associated fingolimod targets (F-ADGs). Immune cell infiltration analysis and a primary mouse cortical culture RNA-seq drug screen database were used to identify immune-related F-ADGs and cortex-related F-ADGs. Then, the expression values of F-ADGs were correlated with the disease severity score (MMSE score) of AD patients to identify severity-related F-ADGs. We also analyzed miRNA expression microarray data in the frontal cortex of AD patients associated with disease severity to obtain severity-related F-ADG-miRNAs. Results: A total of 188 F-ADGs were detected in the frontal cortices of AD patients and were enriched in biological processes such as synaptic signaling, inflammatory response, and response to oxygen-containing compounds. Eleven immune-related F-ADGs (like FPR1, BLNK.) and 17 cortex-related F-ADGs (like ALDH1L1, DUSP1.) were detected. Other F-ADGs, such as S1PR1 and GABBR2, although not classified into the above two categories, were still predicted by bioinformatics methods to play an important role in the development of AD. Two F-ADGs (GNAQ and MMP14) and 28 miRNAs (like miR- 323a-3p, miR-181a-5p.) were found to be associated with AD severity (MMSE 0-27 group). Fifteen F-ADGs (like ALDH1L1, FPR1, and IL6.) and 46 miRNAs (like miR-212-5p, miR-93-5p.) were found to be associated with mild or moderate dementia AD patients' severity (MMSE11-22 subgroup). Conclusions: Fingolimod may affect the brain frontal cortex function of AD patients in many different ways, such as affecting immune cell infiltration, nerve cell, or glial cell function, and synaptic function. miRNAs may also be involved. ALDH1L1, FPR1, S1PR1, and GABBR2 may be core drug targets.
Collapse
Affiliation(s)
- Pengqi Yin
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Xue
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tingting Wang
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States.,Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - Di Zhong
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guozhong Li
- Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
172
|
D’yakonov VA, Dzhemileva LU, Dzhemilev UM. Natural compounds with bis-methylene-interrupted Z-double bonds: plant sources, strategies of total synthesis, biological activity, and perspectives. PHYTOCHEMISTRY REVIEWS 2021; 20:325-342. [DOI: 10.1007/s11101-020-09685-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 05/19/2020] [Indexed: 01/03/2025]
|
173
|
Nikolac Perkovic M, Videtic Paska A, Konjevod M, Kouter K, Svob Strac D, Nedic Erjavec G, Pivac N. Epigenetics of Alzheimer's Disease. Biomolecules 2021; 11:195. [PMID: 33573255 PMCID: PMC7911414 DOI: 10.3390/biom11020195] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/19/2021] [Accepted: 01/26/2021] [Indexed: 02/07/2023] Open
Abstract
There are currently no validated biomarkers which can be used to accurately diagnose Alzheimer's disease (AD) or to distinguish it from other dementia-causing neuropathologies. Moreover, to date, only symptomatic treatments exist for this progressive neurodegenerative disorder. In the search for new, more reliable biomarkers and potential therapeutic options, epigenetic modifications have emerged as important players in the pathogenesis of AD. The aim of the article was to provide a brief overview of the current knowledge regarding the role of epigenetics (including mitoepigenetics) in AD, and the possibility of applying these advances for future AD therapy. Extensive research has suggested an important role of DNA methylation and hydroxymethylation, histone posttranslational modifications, and non-coding RNA regulation (with the emphasis on microRNAs) in the course and development of AD. Recent studies also indicated mitochondrial DNA (mtDNA) as an interesting biomarker of AD, since dysfunctions in the mitochondria and lower mtDNA copy number have been associated with AD pathophysiology. The current evidence suggests that epigenetic changes can be successfully detected, not only in the central nervous system, but also in the cerebrospinal fluid and on the periphery, contributing further to their potential as both biomarkers and therapeutic targets in AD.
Collapse
Affiliation(s)
- Matea Nikolac Perkovic
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Ruder Boskovic Institute, HR-10000 Zagreb, Croatia; (M.N.P.); (M.K.); (D.S.S.); (G.N.E.)
| | - Alja Videtic Paska
- Medical Center for Molecular Biology, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia; (A.V.P.); (K.K.)
| | - Marcela Konjevod
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Ruder Boskovic Institute, HR-10000 Zagreb, Croatia; (M.N.P.); (M.K.); (D.S.S.); (G.N.E.)
| | - Katarina Kouter
- Medical Center for Molecular Biology, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia; (A.V.P.); (K.K.)
| | - Dubravka Svob Strac
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Ruder Boskovic Institute, HR-10000 Zagreb, Croatia; (M.N.P.); (M.K.); (D.S.S.); (G.N.E.)
| | - Gordana Nedic Erjavec
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Ruder Boskovic Institute, HR-10000 Zagreb, Croatia; (M.N.P.); (M.K.); (D.S.S.); (G.N.E.)
| | - Nela Pivac
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Ruder Boskovic Institute, HR-10000 Zagreb, Croatia; (M.N.P.); (M.K.); (D.S.S.); (G.N.E.)
| |
Collapse
|
174
|
Chávez-Fumagalli MA, Shrivastava P, Aguilar-Pineda JA, Nieto-Montesinos R, Del-Carpio GD, Peralta-Mestas A, Caracela-Zeballos C, Valdez-Lazo G, Fernandez-Macedo V, Pino-Figueroa A, Vera-Lopez KJ, Lino Cardenas CL. Diagnosis of Alzheimer's Disease in Developed and Developing Countries: Systematic Review and Meta-Analysis of Diagnostic Test Accuracy. J Alzheimers Dis Rep 2021; 5:15-30. [PMID: 33681713 PMCID: PMC7902992 DOI: 10.3233/adr-200263] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The present systematic review and meta-analysis of diagnostic test accuracy summarizes the last three decades in advances on diagnosis of Alzheimer's disease (AD) in developed and developing countries. OBJECTIVE To determine the accuracy of biomarkers in diagnostic tools in AD, for example, cerebrospinal fluid, positron emission tomography (PET), and magnetic resonance imaging (MRI), etc. METHODS The authors searched PubMed for published studies from 1990 to April 2020 on AD diagnostic biomarkers. 84 published studies were pooled and analyzed in this meta-analysis and diagnostic accuracy was compared by summary receiver operating characteristic statistics. RESULTS Overall, 84 studies met the criteria and were included in a meta-analysis. For EEG, the sensitivity ranged from 67 to 98%, with a median of 80%, 95% CI [75, 91], tau-PET diagnosis sensitivity ranged from 76 to 97%, with a median of 94%, 95% CI [76, 97]; and MRI sensitivity ranged from 41 to 99%, with a median of 84%, 95% CI [81, 87]. Our results showed that tau-PET diagnosis had higher performance as compared to other diagnostic methods in this meta-analysis. CONCLUSION Our findings showed an important discrepancy in diagnostic data for AD between developed and developing countries, which can impact global prevalence estimation and management of AD. Also, our analysis found a better performance for the tau-PET diagnostic over other methods to diagnose AD patients, but the expense of tau-PET scan seems to be the limiting factor in the diagnosis of AD in developing countries such as those found in Asia, Africa, and Latin America.
Collapse
Affiliation(s)
- Miguel A. Chávez-Fumagalli
- Laboratory of Genomics and Neurovascular Diseases, Vicerrectorado de investigación, Universidad Católica de Santa Maria, Arequipa, Peru
| | - Pallavi Shrivastava
- Laboratory of Genomics and Neurovascular Diseases, Vicerrectorado de investigación, Universidad Católica de Santa Maria, Arequipa, Peru
| | - Jorge A. Aguilar-Pineda
- Laboratory of Genomics and Neurovascular Diseases, Vicerrectorado de investigación, Universidad Católica de Santa Maria, Arequipa, Peru
| | - Rita Nieto-Montesinos
- Laboratory of Genomics and Neurovascular Diseases, Vicerrectorado de investigación, Universidad Católica de Santa Maria, Arequipa, Peru
| | - Gonzalo Davila Del-Carpio
- Laboratory of Genomics and Neurovascular Diseases, Vicerrectorado de investigación, Universidad Católica de Santa Maria, Arequipa, Peru
| | - Antero Peralta-Mestas
- Division of Neurology, Psychiatry and Radiology of the National Hospital ESSALUD-HNCASE, Arequipa, Peru
| | - Claudia Caracela-Zeballos
- Division of Neurology, Psychiatry and Radiology of the National Hospital ESSALUD-HNCASE, Arequipa, Peru
| | - Guillermo Valdez-Lazo
- Division of Neurology, Psychiatry and Radiology of the National Hospital ESSALUD-HNCASE, Arequipa, Peru
| | - Victor Fernandez-Macedo
- Division of Neurology, Psychiatry and Radiology of the National Hospital ESSALUD-HNCASE, Arequipa, Peru
| | - Alejandro Pino-Figueroa
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA
| | - Karin J. Vera-Lopez
- Laboratory of Genomics and Neurovascular Diseases, Vicerrectorado de investigación, Universidad Católica de Santa Maria, Arequipa, Peru
| | - Christian L. Lino Cardenas
- Laboratory of Genomics and Neurovascular Diseases, Vicerrectorado de investigación, Universidad Católica de Santa Maria, Arequipa, Peru
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
175
|
Lu Y, Liu C, Yu D, Fawkes S, Ma J, Zhang M, Li C. Prevalence of mild cognitive impairment in community-dwelling Chinese populations aged over 55 years: a meta-analysis and systematic review. BMC Geriatr 2021; 21:10. [PMID: 33407219 PMCID: PMC7789349 DOI: 10.1186/s12877-020-01948-3] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 12/06/2020] [Indexed: 12/16/2022] Open
Abstract
Background Mild cognitive impairment (MCI) is an intermediate phase between normal cognitive ageing and overt dementia, with amnesic MCI (aMCI) being the dominant subtype. This study aims to synthesise the prevalence results of MCI and aMCI in community-dwelling populations in China through a meta-analysis and systematic review. Methods The study followed the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) protocol. English and Chinese studies published before 1 March 2020 were searched from ten electronic bibliographic databases. Two reviewers screened for relevance of the studies against the pre-defined inclusion and exclusion criteria and assessed the quality of the included studies using the Risk of Bias Tool independently. A random-effect model was adopted to estimate the prevalence of MCI and aMCI, followed by sub-group analyses and meta-regression. Sensitivity and publication bias tests were performed to verify the robustness of the meta-analyses. Results A total of 41 studies with 112,632 participants were included in the meta-analyses. The Chinese community-dwelling populations over 55 years old had a pooled prevalence of 12.2% [95% confidence interval (CI): 10.6, 14.2%] for MCI and 10.9% [95% CI, 7.7, 15.4%] for aMCI, respectively. The prevalence of MCI increased with age. The American Psychiatric Association’s Diagnostic tool (DSM-IV) generated the highest MCI prevalence (13.5%), followed by the Petersen criteria (12.9%), and the National Institute on Aging Alzheimer’s Association (NIA-AA) criteria (10.3%). Women, rural residents, and those who lived alone and had low levels of education had higher MCI prevalence than others. Conclusion Higher MCI prevalence was identified in community-dwelling older adult populations in China compared with some other countries, possibly due to more broadened criteria being adopted for confirming the diagnosis. The study shows that aMCI accounts for 66.5% of MCI, which is consistent with findings of studies undertaken elsewhere. Systematic review registration number PROSPERO CRD42019134686. Supplementary Information The online version contains supplementary material available at 10.1186/s12877-020-01948-3.
Collapse
Affiliation(s)
- Yuan Lu
- Department of General Practice, Yangpu hospital, Tongji University School of Medicine, Shanghai, 200090, China.,School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia.,Academic Department of General Practice, Yangpu hospital, Tongji University School of Medicine, Shanghai, 200090, China.,Shanghai General Practice and Community Health Development Research Center, 200090, Shanghai, China
| | - Chaojie Liu
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia.
| | - Dehua Yu
- Department of General Practice, Yangpu hospital, Tongji University School of Medicine, Shanghai, 200090, China. .,Academic Department of General Practice, Yangpu hospital, Tongji University School of Medicine, Shanghai, 200090, China. .,Shanghai General Practice and Community Health Development Research Center, 200090, Shanghai, China.
| | - Sally Fawkes
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Jia Ma
- Academic Department of General Practice, Yangpu hospital, Tongji University School of Medicine, Shanghai, 200090, China
| | - Min Zhang
- Academic Department of General Practice, Yangpu hospital, Tongji University School of Medicine, Shanghai, 200090, China
| | - Chunbo Li
- Clinical Research Center, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shangha, China
| |
Collapse
|
176
|
Amiri A, Barreto G, Sathyapalan T, Sahebkar A. siRNA Therapeutics: Future Promise for Neurodegenerative Diseases. Curr Neuropharmacol 2021; 19:1896-1911. [PMID: 33797386 PMCID: PMC9185778 DOI: 10.2174/1570159x19666210402104054] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/29/2021] [Accepted: 03/30/2021] [Indexed: 11/22/2022] Open
Abstract
Neurodegenerative diseases (ND), as a group of central nervous system (CNS) disorders, are among the most prominent medical problems of the 21st century. They are often associated with considerable disability, motor dysfunction and dementia and are more common in the aged population. ND imposes a psychologic, economic and social burden on the patients and their families. Currently, there is no effective treatment for ND. Since many ND result from the gain of function of a mutant allele, small interference RNA (siRNA) can be a potential therapeutic agent for ND management. Based on the RNA interference (RNAi) approach, siRNA is a powerful tool for modulating gene expression through gene silencing. However, there are some obstacles in the clinical application of siRNA, including unfavorable immune response, off-target effects, instability of naked siRNA, nuclease susceptibility and a need to develop a suitable delivery system. Since there are some issues related to siRNA delivery routes, in this review, we focus on the application of siRNA in the management of ND treatment from 2000 to 2020.
Collapse
Affiliation(s)
| | | | | | - Amirhossein Sahebkar
- Address correspondence to this author at the Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Tel: 985118002288; Fax: 985118002287; E-mails: ;
| |
Collapse
|
177
|
Guo X, Wang Y, Li Y, Liu Y, Liu Y, Chen D, Xiao J, Gao W, Zhou B, Liu Y, Liu R, Chen W, Liu F, Guo D, Mao G, Huang H. A pilot study of clinical cell therapies in Alzheimer’s disease. JOURNAL OF NEURORESTORATOLOGY 2021. [DOI: 10.26599/jnr.2021.9040023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease dominated by progressive cognitive dysfunction causing significant social, economic, and medical crises. Cell therapy has demonstrated favorable effects for AD. This pilot study examined the safety and neurorestorative effects of the olfactory ensheathing cell (OEC), olfactory neuron (ON), and Schwann cell (SC) on patients with AD. Seven patients with AD were enrolled in this two-center, randomized, double-blind, and placebo- controlled cell therapy study with a subsequent 12-month follow-up. We randomly assigned one or two participants in OEC, ON, and SC therapy or OEC combined with ON and placebo control. All enrolled patients were injected cells or medium into the olfactory sub-mucosa. They got an assessment of Mini-Mental State Examination, Montreal Cognitive Assessment, and Clinical Dementia Rating before treatment and 1, 3, 6, 12 months after treatment. We performed MRI or CT scans before treatment and 12 months after treatment. After integrating the results from the three evaluation methods, all cell types showed better results than a placebo control. ON and SC seem to exhibit more vital potential neurorestorative ability to enhance or convert the neurological functions of patients with AD, and OEC may help AD patients keep neurological functions stable. In this pilot study, there was no adverse or side-effect event. The results of this study strongly suggest conducting a phase II clinical trial of ON, SC, and OEC therapy to prove their neurorestorative effect on patients with AD.
Collapse
|
178
|
Hughes TF, Liu A, Jacobsen E, Rosano C, Berman SB, Chang CCH, Ganguli M. Exercise and the Risk of Mild Cognitive Impairment: Does the Effect Depend on Vascular Factors? Alzheimer Dis Assoc Disord 2021; 35:30-35. [PMID: 32960854 PMCID: PMC9350907 DOI: 10.1097/wad.0000000000000410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Although exercise is associated with a lower risk for mild cognitive impairment (MCI), it is unclear whether its protective effect depends on the presence or absence of vascular factors. METHODS In an exploratory study of data from a population-based cohort, 1254 participants aged 65+ years were followed for 10 years for incident MCI. The main effect of baseline total minutes of exercise per week (0 vs. 1 to 149 vs. 150+), and its interaction with several vascular factors, on risk for incident MCI was examined using Cox proportional hazards regression models, adjusting for demographics. RESULTS Compared with no exercise, 1 to 149 minutes [hazard ratio (HR)=0.90; 95% confidence interval (95% CI), 0.69-1.16] and 150 or more minutes per week (HR=0.84; 95% CI, 0.66-1.07) of exercise lowered risk for incident MCI in a dose-dependent manner. The majority of interactions were not statistically significant, but risk reduction effect sizes of <0.75 suggested that exercise may have stronger effects among those without high cholesterol, never smoking, and not currently consuming alcohol; also, those with arrhythmia, coronary artery disease, and heart failure. Overall, there was a pattern of exercise being associated with lower MCI risk among those without vascular factors. CONCLUSIONS Spending more time engaging in exercise each week may offer protection against MCI in late life, with some variation among those with different vascular conditions and risk factors. Our findings may help target subgroups for exercise recommendations and interventions, and also generate hypotheses to test regarding underlying mechanisms.
Collapse
Affiliation(s)
- Tiffany F. Hughes
- Youngstown State University, Youngstown, OH, United States of America
| | - Anran Liu
- University of Pittsburgh, Graduate School of Public Health, Department of Biostatistics, Pittsburgh, PA, United States of America
| | - Erin Jacobsen
- University of Pittsburgh, School of Medicine, Department of Psychiatry, Pittsburgh, PA, United States of America
| | - Caterina Rosano
- University of Pittsburgh, Graduate School of Public Health, Department of Epidemiology, Pittsburgh, PA, United State of America
| | - Sarah B. Berman
- University of Pittsburgh, School of Medicine, Department of Neurology, Pittsburgh, PA, United States of America
| | - Chung-Chou H. Chang
- University of Pittsburgh, Graduate School of Public Health, Department of Biostatistics, Pittsburgh, PA, United States of America
- University of Pittsburgh, School of Medicine, Department of Medicine, Pittsburgh, PA, United States of America
| | - Mary Ganguli
- University of Pittsburgh, School of Medicine, Department of Psychiatry, Pittsburgh, PA, United States of America
- University of Pittsburgh, Graduate School of Public Health, Department of Epidemiology, Pittsburgh, PA, United State of America
- University of Pittsburgh, School of Medicine, Department of Neurology, Pittsburgh, PA, United States of America
| |
Collapse
|
179
|
Farfel JM, Barnes LL, Capuano A, de Moraes Sampaio MC, Wilson RS, Bennett DA. Informant-Reported Discrimination, Dementia, and Cognitive Impairment in Older Brazilians. J Alzheimers Dis 2021; 84:973-981. [PMID: 33935076 PMCID: PMC9113828 DOI: 10.3233/jad-201436] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Self-reported discrimination is a source of psychosocial stress that has been previously associated with poor cognitive function in older African Americans without dementia. OBJECTIVE Here, we examine the association of discrimination with dementia and cognitive impairment in racially diverse older Brazilians. METHODS We included 899 participants 65 years or older (34.3% Black) from the Pathology, Alzheimer's and Related Dementias Study (PARDoS), a community-based study of aging and dementia. A structured interview with informants of the deceased was conducted. The interview included the Clinical Dementia Rating (CDR) Scale for the diagnosis of dementia and cognitive impairment proximate to death and the Informant Questionnaire on Cognitive Decline in the Elderly (IQCODE) as a second measure of cognitive impairment. Informant-reported discrimination was assessed using modified items from the Major and Everyday Discrimination Scales. RESULTS Discrimination was reported by informants of 182 (20.2%) decedents and was more likely reported by informants of Blacks than Whites (25.3% versus 17.6%, p = 0.006). Using the CDR, a higher level of informant-reported discrimination was associated with higher odds of dementia (OR: 1.24, 95% CI 1.08 -1.42, p = 0.002) and cognitive impairment (OR: 1.21, 95% CI: 1.06 -1.39, p = 0.004). Similar results were observed using the IQCODE (estimate: 0.07, SE: 0.02, p = 0.003). The effects were independent of race, sex, education, socioeconomic status, major depression, neuroticism, or comorbidities. CONCLUSION Higher level of informant-reported discrimination was associated with higher odds of dementia and cognitive impairment in racially diverse older Brazilians.
Collapse
Affiliation(s)
- Jose M. Farfel
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Pathology, Rush University Medical Center, Chicago, IL, USA
- Health Sciences Program, Instituto de Assistência Medica ao Servidor Público do Estado (IAMSPE), São Paulo, Brazil
| | - Lisa L. Barnes
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush Medical College, Chicago, IL, USA
- Department of Psychiatry and Behavioral Sciences, Rush Medical College, Chicago, IL, USA
| | - Ana Capuano
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Health Sciences Program, Instituto de Assistência Medica ao Servidor Público do Estado (IAMSPE), São Paulo, Brazil
- Department of Neurological Sciences, Rush Medical College, Chicago, IL, USA
| | | | - Robert S. Wilson
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Neurological Sciences, Rush Medical College, Chicago, IL, USA
- Department of Psychiatry and Behavioral Sciences, Rush Medical College, Chicago, IL, USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
- Health Sciences Program, Instituto de Assistência Medica ao Servidor Público do Estado (IAMSPE), São Paulo, Brazil
- Department of Neurological Sciences, Rush Medical College, Chicago, IL, USA
| |
Collapse
|
180
|
Hassan H, Chen R. Hypoxia in Alzheimer's disease: effects of hypoxia inducible factors. Neural Regen Res 2021; 16:310-311. [PMID: 32859789 PMCID: PMC7896215 DOI: 10.4103/1673-5374.290898] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Halimatu Hassan
- School of Pharmacy and Bioengineering, Keele University, Staffordshire, UK
| | - Ruoli Chen
- School of Pharmacy and Bioengineering, Keele University, Staffordshire, UK
| |
Collapse
|
181
|
Williams VJ, Carlsson CM, Fischer A, Johnson SC, Lange K, Partridge E, Roan C, Asthana S, Herd P. Assessing Dementia Prevalence in the Wisconsin Longitudinal Study: Cohort Profile, Protocol, and Preliminary Findings. J Alzheimers Dis 2021; 81:751-768. [PMID: 33843672 PMCID: PMC10551824 DOI: 10.3233/jad-201422] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND There is growing consensus that non-genetic determinants of dementia can be linked to various risk- and resiliency-enhancing factors accumulating throughout the lifespan, including socioeconomic conditions, early life experiences, educational attainment, lifestyle behaviors, and physical/mental health. Yet, the causal impact of these diverse factors on dementia risk remain poorly understood due to few longitudinal studies prospectively characterizing these influences across the lifespan. OBJECTIVE The Initial Lifespan's Impact on Alzheimer's Disease and Related Dementia (ILIAD) study aims to characterize dementia prevalence in the Wisconsin Longitudinal Study (WLS), a 60-year longitudinal study documenting life course trajectories of educational, family, occupational, psychological, cognitive, and health measures. METHODS Participants are surveyed using the modified Telephone Interview for Cognitive Status (TICS-m) to identify dementia risk. Those scoring below cutoff undergo home-based neuropsychological, physical/neurological, and functional assessments. Dementia diagnosis is determined by consensus panel and merged with existing WLS data for combined analysis. RESULTS Preliminary findings demonstrate the initial success of the ILIAD protocol in detecting dementia prevalence in the WLS. Increasing age, hearing issues, lower IQ, male sex, APOE4 positivity, and a steeper annualized rate of memory decline assessed in the prior two study waves, all increased likelihood of falling below the TICS-m cutoff for dementia risk. TICS-m scores significantly correlated with standard neuropsychological performance and functional outcomes. CONCLUSION We provide an overview of the WLS study, describe existing key lifespan variables relevant to studies of dementia and cognitive aging, detail the current WLS-ILIAD study protocol, and provide a first glimpse of preliminary study findings.
Collapse
Affiliation(s)
- Victoria J. Williams
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Cynthia M. Carlsson
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research, Education, and Clinical Center (GRECC), William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Anne Fischer
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Sterling C. Johnson
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research, Education, and Clinical Center (GRECC), William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Kate Lange
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Eileen Partridge
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
| | - Carol Roan
- Department of Sociology, University of Wisconsin at Madison, Department of Sociology, Madison, WI, USA
| | - Sanjay Asthana
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research, Education, and Clinical Center (GRECC), William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Pamela Herd
- McCourt School of Public Policy, Georgetown University, Washington, DC, USA
| |
Collapse
|
182
|
Snyder PJ, Alber J, Alt C, Bain LJ, Bouma BE, Bouwman FH, DeBuc DC, Campbell MC, Carrillo MC, Chew EY, Cordeiro MF, Dueñas MR, Fernández BM, Koronyo-Hamaoui M, La Morgia C, Carare RO, Sadda SR, van Wijngaarden P, Snyder HM. Retinal imaging in Alzheimer's and neurodegenerative diseases. Alzheimers Dement 2021; 17:103-111. [PMID: 33090722 PMCID: PMC8062064 DOI: 10.1002/alz.12179] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/26/2020] [Accepted: 07/27/2020] [Indexed: 12/22/2022]
Abstract
In the last 20 years, research focused on developing retinal imaging as a source of potential biomarkers for Alzheimer's disease and other neurodegenerative diseases, has increased significantly. The Alzheimer's Association and the Alzheimer's & Dementia: Diagnosis, Assessment, Disease Monitoring editorial team (companion journal to Alzheimer's & Dementia) convened an interdisciplinary discussion in 2019 to identify a path to expedite the development of retinal biomarkers capable of identifying biological changes associated with AD, and for tracking progression of disease severity over time. As different retinal imaging modalities provide different types of structural and/or functional information, the discussion reflected on these modalities and their respective strengths and weaknesses. Discussion further focused on the importance of defining the context of use to help guide the development of retinal biomarkers. Moving from research to context of use, and ultimately to clinical evaluation, this article outlines ongoing retinal imaging research today in Alzheimer's and other brain diseases, including a discussion of future directions for this area of study.
Collapse
Affiliation(s)
- Peter J. Snyder
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island
| | - Jessica Alber
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island
| | - Clemens Alt
- Wellman Center for Photomedicine and Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lisa J. Bain
- Independent Science Writer, Elverson, Pennsylvania
| | - Brett E. Bouma
- Harvard Medical School, Massachusetts General Hospital and Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Massachusetts
| | - Femke H. Bouwman
- Neurologist, Alzheimer Center Amsterdam UMC, Amsterdam, The Netherlands
| | | | - Melanie C.W. Campbell
- Physics and Astronomy, Optometry and Vision Science and Systems Design Engineering, University of Waterloo, Waterloo, Ontario, Canada
| | - Maria C. Carrillo
- Medical & Scientific Relations, Alzheimer’s Association, Chicago, Illinois
| | - Emily Y. Chew
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - M. Francesca Cordeiro
- Imperial College London, UCL Institute of Ophthalmology, ICORG Western Eye Hospital, London, UK
| | - Michael R. Dueñas
- Chief Public Health Officer (Ret.), American Optometric Association, Washington, D.C
| | | | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute and Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Chiara La Morgia
- IRCCS Istituto delle Scienze Neurologiche di Bologna, UOC Clinica Neurologica, Bologna, Italy
- Dipartimento di Scienze Biomediche e Neuromotorie, University of Bologna, Italy
| | | | - Srinivas R. Sadda
- Doheny Eye Institute, Los Angeles, California
- Department of Ophthalmology, UCLA, Los Angeles, California
| | - Peter van Wijngaarden
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, Parkville, Australia
| | - Heather M. Snyder
- Medical & Scientific Relations, Alzheimer’s Association, Chicago, Illinois
| |
Collapse
|
183
|
Moradi S, Moloudi J, Moradinazar M, Sarokhani D, Nachvak SM, Samadi M. Adherence to Healthy Diet Can Delay Alzheimer's Diseases Development: A Systematic Review and Meta-Analysis. Prev Nutr Food Sci 2020; 25:325-337. [PMID: 33505926 PMCID: PMC7813603 DOI: 10.3746/pnf.2020.25.4.325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/15/2020] [Indexed: 11/07/2022] Open
Abstract
A healthy diet has long been indicated to be protective against Alzheimer’s diseases (AD). We carried out a systematic review and meta-analysis of published observational studies to explore the relationship between healthy and unhealthy diets and risk of ADs. We screened PubMed, Scopus, Web of Sciences, Google Scholar, Science Direct, and Embase, and screened manually to identify relevant articles published in English and non-English until Jun 2020. We classified the studied dietary patterns into two groups: healthy and unhealthy diets. The pooled weighted mean difference and 95% confidence interval (95% CI) was used to analyze the data using a random-effects model. The data were extracted manually and the preferred reporting items for systematic review and meta-analysis checklist was used to appraise the risk of bias and quality of data. Of the 1,813 articles identified, 21 met the inclusion criteria and were included in the quantitative analysis. A healthy diet was related to a lower risk of AD [odds ratio (OR): 0.45, 95% CI: 0.23 to 0.86, I2=99.7%; n=17 studies]. Moreover, high adherence to an unhealthy diet was not associated with increased risk of AD (OR: 0.99, 95% CI: 0.98 to 0.99, I2=0.0%; n=6 studies). However, the etiology of AD is uncertain and it is difficult draw conclusions about dietary healthy patterns. We concluded that adherence to a healthy diet is associated with a lower risk of AD, but were unable to find evidence that an unhealthy diet increases the risk of AD.
Collapse
Affiliation(s)
- Shima Moradi
- Department of Nutritional Sciences, Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah 67198-51351, Iran
| | - Jalall Moloudi
- Department of Nutritional Sciences, Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah 67198-51351, Iran.,Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah 67198-51351, Iran
| | - Mehdi Moradinazar
- Behavioral Disease Research Center, Kermanshah University of Medical Sciences, Kermanshah 67198-51351, Iran
| | - Diana Sarokhani
- Psychosocial Injuries Research Center, Ilam University of Medical Sciences, Ilam 69311-57793, Iran
| | - Seyyed Mostafa Nachvak
- Department of Nutritional Sciences, Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah 67198-51351, Iran
| | - Mehnoosh Samadi
- Department of Nutritional Sciences, Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah 67198-51351, Iran
| |
Collapse
|
184
|
Physical Therapy for Gait, Balance, and Cognition in Individuals with Cognitive Impairment: A Retrospective Analysis. Rehabil Res Pract 2020; 2020:8861004. [PMID: 33204533 PMCID: PMC7655244 DOI: 10.1155/2020/8861004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/08/2020] [Accepted: 10/17/2020] [Indexed: 11/29/2022] Open
Abstract
Objectives The purpose of this study was to determine if a pragmatic physical therapy (PT) program was associated with improved cognition, gait, and balance in individuals with cognitive impairment. This study investigated these associations for individuals with Alzheimer disease (AD), vascular dementia (VaD), dementia with Lewy bodies (DLB), and mild cognitive impairment (MCI) in order to better characterize outcomes to PT for each diagnostic group. Methods Data before and after one month of physical therapy were extracted from patient records (67 with AD, 34 with VaD, 35 with DLB, and 37 with MCI). The mean number of PT sessions over a month was 3.4 (±1.8). Outcomes covered the domains of gait, balance, and cognition with multiple outcomes used to measure different constructs within the balance and gait domains. Results All groups showed improvements in balance and at least one gait outcome measure. Those with MCI improved in every measure of gait and balance performance. Lastly, cognition as measured by Montreal Cognitive Assessment improved in individuals in the AD, VaD, and MCI groups. Conclusion While this retrospective analysis is not appropriate for causal inference, results of one month of physical therapy were associated with decreases in gait, balance, and cognitive impairment in individuals with AD, VaD, DLB<, and MCI. Clinical Implications. While physical therapy is not typically a primary treatment strategy for individuals with cognitive impairment, the results of this study are consistent with the literature that demonstrates improvement from physical therapy for other neurodegenerative diseases. Further clinical and research exploration for physical therapy as a primary treatment strategy in these populations is warranted.
Collapse
|
185
|
Self- assembled lactoferrin-conjugated linoleic acid micelles as an orally active targeted nanoplatform for Alzheimer's disease. Int J Biol Macromol 2020; 162:246-261. [DOI: 10.1016/j.ijbiomac.2020.06.058] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/22/2020] [Accepted: 06/07/2020] [Indexed: 12/29/2022]
|
186
|
Alber J, Goldfarb D, Thompson LI, Arthur E, Hernandez K, Cheng D, DeBuc DC, Cordeiro F, Provetti-Cunha L, den Haan J, Van Stavern GP, Salloway SP, Sinoff S, Snyder PJ. Developing retinal biomarkers for the earliest stages of Alzheimer's disease: What we know, what we don't, and how to move forward. Alzheimers Dement 2020; 16:229-243. [PMID: 31914225 DOI: 10.1002/alz.12006] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/23/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022]
Abstract
The last decade has seen a substantial increase in research focused on the identification, development, and validation of diagnostic and prognostic retinal biomarkers for Alzheimer's disease (AD). Sensitive retinal biomarkers may be advantageous because they are cost and time efficient, non-invasive, and present a minimal degree of patient risk and a high degree of accessibility. Much of the work in this area thus far has focused on distinguishing between symptomatic AD and/or mild cognitive impairment (MCI) and cognitively normal older adults. Minimal work has been done on the detection of preclinical AD, the earliest stage of AD pathogenesis characterized by the accumulation of cerebral amyloid absent clinical symptoms of MCI or dementia. The following review examines retinal structural changes, proteinopathies, and vascular alterations that have been proposed as potential AD biomarkers, with a focus on studies examining the earliest stages of disease pathogenesis. In addition, we present recommendations for future research to move beyond the discovery phase and toward validation of AD risk biomarkers that could potentially be used as a first step in a multistep screening process for AD risk detection.
Collapse
Affiliation(s)
- Jessica Alber
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island, USA.,George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island, USA.,Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, Rhode Island, USA.,Butler Hospital Memory & Aging Program, Providence, Rhode Island, USA
| | | | - Louisa I Thompson
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, Rhode Island, USA.,Butler Hospital Memory & Aging Program, Providence, Rhode Island, USA
| | - Edmund Arthur
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island, USA.,George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island, USA.,Butler Hospital Memory & Aging Program, Providence, Rhode Island, USA
| | | | - Derrick Cheng
- Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Delia Cabrera DeBuc
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami, Miami, Florida, USA
| | - Francesca Cordeiro
- Imperial College London, London, UK.,University College London, London, UK.,Western Eye Hospital, London, UK
| | - Leonardo Provetti-Cunha
- Federal University of Juiz de Fora Medical School, Juiz de Fora, Minas Gerais, Brazil.,Juiz de Fora Eye Hospital, Juiz de Fora, Minas Gerais, Brazil.,University of São Paulo Medical School, São Paulo, Brazil
| | - Jurre den Haan
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Gregory P Van Stavern
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Stephen P Salloway
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, Rhode Island, USA.,Butler Hospital Memory & Aging Program, Providence, Rhode Island, USA.,Department of Neurology, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | | | - Peter J Snyder
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island, USA.,George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island, USA.,Department of Neurology and Department of Surgery (Ophthalmology), Alpert Medical School of Brown University, Providence, Rhode Island, USA
| |
Collapse
|
187
|
So JH, Madusanka N, Choi HK, Choi BK, Park HG. Deep Learning for Alzheimer's Disease Classification using Texture Features. Curr Med Imaging 2020; 15:689-698. [PMID: 32008517 DOI: 10.2174/1573405615666190404163233] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/08/2019] [Accepted: 03/21/2019] [Indexed: 01/27/2023]
Abstract
BACKGROUND We propose a classification method for Alzheimer's disease (AD) based on the texture of the hippocampus, which is the organ that is most affected by the onset of AD. METHODS We obtained magnetic resonance images (MRIs) of Alzheimer's patients from the Alzheimer's Disease Neuroimaging Initiative (ADNI) dataset. This dataset consists of image data for AD, mild cognitive impairment (MCI), and normal controls (NCs), classified according to the cognitive condition. In this study, the research methods included image processing, texture analyses, and deep learning. Firstly, images were acquired for texture analyses, which were then re-spaced, registered, and cropped with Gabor filters applied to the resulting image data. In the texture analyses, we applied the 3-dimensional (3D) gray-level co-occurrence (GLCM) method to evaluate the textural features of the image, and used Fisher's coefficient to select the appropriate features for classification. In the last stage, we implemented a deep learning multi-layer perceptron (MLP) model, which we divided into three types, namely, AD-MCI, AD-NC, and MCI-NC. RESULTS We used this model to assess the accuracy of the proposed method. The classification accuracy of the proposed deep learning model was confirmed in the cases of AD-MCI (72.5%), ADNC (85%), and MCI-NC (75%). We also evaluated the results obtained using a confusion matrix, support vector machine (SVM), and K-nearest neighbor (KNN) classifier and analyzed the results to objectively verify our model. We obtained the highest accuracy of 85% in the AD-NC. CONCLUSION The proposed model was at least 6-19% more accurate than the SVM and KNN classifiers, respectively. Hence, this study confirms the validity and superiority of the proposed method, which can be used as a diagnostic tool for early Alzheimer's diagnosis.
Collapse
Affiliation(s)
- Jae-Hong So
- Department of Digital Anti-Aging Healthcare, u-AHRC, Inje University, Gimhae, Gyeongsangnam, Korea
| | - Nuwan Madusanka
- Department of Computer Engineering, u-AHRC, Inje University, Gimhae, Gyeongsangnam, Korea
| | - Heung-Kook Choi
- Department of Computer Engineering, u-AHRC, Inje University, Gimhae, Gyeongsangnam, Korea
| | - Boo-Kyeong Choi
- Department of Digital Anti-Aging Healthcare, u-AHRC, Inje University, Gimhae, Gyeongsangnam, Korea
| | - Hyeon-Gyun Park
- Department of Computer Engineering, u-AHRC, Inje University, Gimhae, Gyeongsangnam, Korea
| |
Collapse
|
188
|
Xu J, Wang F, Xu Z, Adekkanattu P, Brandt P, Jiang G, Kiefer RC, Luo Y, Mao C, Pacheco JA, Rasmussen LV, Zhang Y, Isaacson R, Pathak J. Data-driven discovery of probable Alzheimer's disease and related dementia subphenotypes using electronic health records. Learn Health Syst 2020; 4:e10246. [PMID: 33083543 PMCID: PMC7556420 DOI: 10.1002/lrh2.10246] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 07/19/2020] [Accepted: 08/06/2020] [Indexed: 12/04/2022] Open
Abstract
Introduction We sought to assess longitudinal electronic health records (EHRs) using machine learning (ML) methods to computationally derive probable Alzheimer's Disease (AD) and related dementia subphenotypes. Methods A retrospective analysis of EHR data from a cohort of 7587 patients seen at a large, multi‐specialty urban academic medical center in New York was conducted. Subphenotypes were derived using hierarchical clustering from 792 probable AD patients (cases) who had received at least one diagnosis of AD using their clinical data. The other 6795 patients, labeled as controls, were matched on age and gender with the cases and randomly selected in the ratio of 9:1. Prediction models with multiple ML algorithms were trained on this cohort using 5‐fold cross‐validation. XGBoost was used to rank the variable importance. Results Four subphenotypes were computationally derived. Subphenotype A (n = 273; 28.2%) had more patients with cardiovascular diseases; subphenotype B (n = 221; 27.9%) had more patients with mental health illnesses, such as depression and anxiety; patients in subphenotype C (n = 183; 23.1%) were overall older (mean (SD) age, 79.5 (5.4) years) and had the most comorbidities including diabetes, cardiovascular diseases, and mental health disorders; and subphenotype D (n = 115; 14.5%) included patients who took anti‐dementia drugs and had sensory problems, such as deafness and hearing impairment. The 0‐year prediction model for AD risk achieved an area under the receiver operating curve (AUC) of 0.764 (SD: 0.02); the 6‐month model, 0.751 (SD: 0.02); the 1‐year model, 0.752 (SD: 0.02); the 2‐year model, 0.749 (SD: 0.03); and the 3‐year model, 0.735 (SD: 0.03), respectively. Based on variable importance, the top‐ranked comorbidities included depression, stroke/transient ischemic attack, hypertension, anxiety, mobility impairments, and atrial fibrillation. The top‐ranked medications included anti‐dementia drugs, antipsychotics, antiepileptics, and antidepressants. Conclusions Four subphenotypes were computationally derived that correlated with cardiovascular diseases and mental health illnesses. ML algorithms based on patient demographics, diagnosis, and treatment demonstrated promising results in predicting the risk of developing AD at different time points across an individual's lifespan.
Collapse
Affiliation(s)
- Jie Xu
- Department of Population Health Sciences Information Technologies and Services, Weill Cornell Medicine New York New York USA
| | - Fei Wang
- Department of Population Health Sciences Information Technologies and Services, Weill Cornell Medicine New York New York USA
| | - Zhenxing Xu
- Department of Population Health Sciences Information Technologies and Services, Weill Cornell Medicine New York New York USA
| | - Prakash Adekkanattu
- Information Technologies and Services, Weill Cornell Medicine New York New York USA
| | - Pascal Brandt
- Biomedical Informatics and Medical Education University of Washington Seattle Washington USA
| | - Guoqian Jiang
- Department of Health Sciences Research Mayo Clinic Rochester Minnesota USA
| | - Richard C Kiefer
- Department of Health Sciences Research Mayo Clinic Rochester Minnesota USA
| | - Yuan Luo
- Feinberg School of Medicine Northwestern University Chicago Illinois USA
| | - Chengsheng Mao
- Feinberg School of Medicine Northwestern University Chicago Illinois USA
| | - Jennifer A Pacheco
- Feinberg School of Medicine Northwestern University Chicago Illinois USA
| | - Luke V Rasmussen
- Feinberg School of Medicine Northwestern University Chicago Illinois USA
| | - Yiye Zhang
- Department of Population Health Sciences Information Technologies and Services, Weill Cornell Medicine New York New York USA
| | - Richard Isaacson
- Department of Population Health Sciences Information Technologies and Services, Weill Cornell Medicine New York New York USA
| | - Jyotishman Pathak
- Department of Population Health Sciences Information Technologies and Services, Weill Cornell Medicine New York New York USA
| |
Collapse
|
189
|
Association of Late Life Depression, (Non-) Modifiable Risk and Protective Factors with Dementia and Alzheimer's Disease: Literature Review on Current Evidences, Preventive Interventions and Possible Future Trends in Prevention and Treatment of Dementia. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17207475. [PMID: 33066592 PMCID: PMC7602449 DOI: 10.3390/ijerph17207475] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/04/2020] [Accepted: 10/06/2020] [Indexed: 02/07/2023]
Abstract
The number of people living with dementia and Alzheimer’s disease is growing rapidly, making dementia one of the biggest challenges for this century. Many studies have indicated that depression plays an important role in development of dementia, including Alzheimer’s disease; depression, especially, during the late life may either increase the risk of dementia or even being its prodromal stage. Despite a notably large number of carried observational studies and/or clinical trials, the association between the late life depression and dementia remains, due to the complexity of their relationship, still unclear. Moreover, during past two decades multiple other (non-)modifiable risk and possibly protective factors such as the hypertension, social engagement, obesity, level of education or physical (in)activity have been identified and their relationship with the risk for development of dementia and Alzheimer’s disease has been extensively studied. It has been proposed that to understand mechanisms of dementia and Alzheimer’s disease pathogeneses require their multifactorial nature represented by these multiple factors to be considered. In this review, we first summarize the recent literature findings on roles of the late life depression and the other known (non-)modifiable risk and possibly protective factors in development of dementia and Alzheimer’s disease. Then, we provide evidences supporting hypotheses that (i) depressive syndromes in late life may indicate the prodromal stage of dementia (Alzheimer’s disease) and, (ii) the interplay among the multiple (non-)modifiable risk and protective factors should be considered to gain a better understanding of dementia and Alzheimer’s disease pathogeneses. We also discuss the evidences of recently established interventions considered to prevent or delay the prodromes of dementia and provide the prospective future directions in prevention and treatment of dementia and Alzheimer’s disease using both the single-domain and multidomain interventions.
Collapse
|
190
|
Derby CA, Katz MJ, Rozner S, Lipton RB, Hall CB. A Birth Cohort Analysis of Amnestic Mild Cognitive Impairment Incidence in the Einstein Aging Study (EAS) Cohort. J Alzheimers Dis 2020; 70:S271-S281. [PMID: 31256119 PMCID: PMC6700647 DOI: 10.3233/jad-181141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND The transition from normal cognition to Alzheimer's disease is considered a continuum, with amnestic mild cognitive impairment (aMCI) an intermediate clinical cognitive state. Although prior work suggests that dementia incidence rates may be declining, there is little information regarding temporal trends in aMCI incidence. OBJECTIVE To determine whether age specific rates of aMCI have changed over sequential birth cohorts among individuals included in the population-based Einstein Aging Study (EAS) cohort. A secondary objective was to examine trends in aMCI rates among Blacks and Whites and by sex. METHODS Age specific incidence of aMCI was examined by birth year among 1,233 individuals age 70 years and above enrolled in the population-based EAS cohort between November 1, 1993 and February 22, 2016 and who had at least one annual follow-up assessment (5,321 person years of follow-up). Poisson regression was used to determine whether there has been a change in age specific aMCI rates over sequential years of birth. RESULTS No significant change in aMCI rates was identified in the overall cohort, among Blacks or Whites, or among males or females born between 1899 and 1946. CONCLUSIONS Despite a trend for decreased dementia incidence in the EAS cohort, rates of incident aMCI have not changed. These apparently conflicting results may indicate a delay or decrease in the rates of transition from aMCI to dementia within the cohort. However, further studies are needed to confirm whether rates of aMCI have changed in other populations, and how aMCI rates are related to secular trends in dementia risk factors.
Collapse
Affiliation(s)
- Carol A Derby
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mindy J Katz
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sara Rozner
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Richard B Lipton
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Charles B Hall
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
191
|
Greater effect of polygenic risk score for Alzheimer's disease among younger cases who are apolipoprotein E-ε4 carriers. Neurobiol Aging 2020; 99:101.e1-101.e9. [PMID: 33164815 DOI: 10.1016/j.neurobiolaging.2020.09.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 09/04/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022]
Abstract
To evaluate how age and apolipoprotein E-ε4 (APOE4) status interact with APOE-independent polygenic risk score (PRSnon-APOE), we estimated PRSnon-APOE in superagers (age ≥ 90 years, N = 346), 89- controls (age 60-89, N = 2930), and Alzheimer's disease (AD) cases (N = 1760). Using superagers, we see a nearly 5 times greater odds ratio (OR) for AD comparing the top PRSnon-APOE decile to the lowest decile (OR = 4.82, p = 2.5 × 10-6), which is twice the OR as using 89- controls (OR = 2.38, p = 4.6 × 10-9). Thus PRSnon-APOE is correlated with age, which in turn is associated with APOE. Further exploring these relationships, we find that PRSnon-APOE modifies age at onset among APOE4 carriers, but not among noncarriers. More specifically, PRSnon-APOE in the top decile predicts an age at onset 5 years earlier compared with the lowest decile (70.1 vs. 75.0 years; t-test p = 2.4 × 10-5) among APOE4 carriers. This disproportionally large PRSnon-APOE among younger APOE4-positive cases is reflected in a significant statistical interaction between APOE4 status and age at onset (β = -0.02, p = 4.8 × 10-3) as a predictor of PRSnon-APOE. Thus, the known AD risk variants are particularly detrimental in young APOE4 carriers.
Collapse
|
192
|
Impact of benzodiazepine consumption reduction on future burden of dementia. Sci Rep 2020; 10:14666. [PMID: 32887900 PMCID: PMC7474098 DOI: 10.1038/s41598-020-71482-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 07/30/2020] [Indexed: 11/08/2022] Open
Abstract
Dementia is a major public health issue worldwide and chronic use of benzodiazepine, which is very frequent in northern countries, was found to be a risk factor of dementia. This work aims at evaluating the impact of a reduction in chronic use of benzodiazepine on the future burden of dementia in France. Using estimations of dementia incidence and of benzodiazepine use and nation-wide projections of mortality and population sizes, a Monte Carlo approach based on an illness-death model provided projections of several indicators of dementia burden. With no change in benzodiazepine consumption, the prevalence of dementia between age 65 and 99 in France in 2040 was estimated at 2.16 millions (95% confidence interval (CI) 1.93-2.38), with a life expectancy without dementia at 65 years equal to 25.0 years (24.7-25.3) for women and 23.8 years (23.5-24.2) for men. Assuming a disappearance of chronic use of benzodiazepine in 2020, the prevalence would be reduced by about 6.6% in 2040 and the life expectancy without dementia would increase by 0.99 (0.93-1.06) year among women and 0.56 (0.50-0.62) among men. To conclude, a modest but significant reduction in future dementia burden could be obtained by applying current recommendation for duration of benzodiazepine use.
Collapse
|
193
|
Hayes-Larson E, Ackley SF, Zimmerman SC, Ospina-Romero M, Glymour MM, Graff RE, Witte JS, Kobayashi LC, Mayeda ER. The competing risk of death and selective survival cannot fully explain the inverse cancer-dementia association. Alzheimers Dement 2020; 16:1696-1703. [PMID: 32881307 DOI: 10.1002/alz.12168] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/16/2020] [Accepted: 07/09/2020] [Indexed: 12/14/2022]
Abstract
INTRODUCTION We evaluated whether competing risk of death or selective survival could explain the reported inverse association between cancer history and dementia incidence (incidence rate ratio [IRR] ≈ 0.62-0.85). METHODS A multistate simulation model of a cancer- and dementia-free cohort of 65-year-olds was parameterized with real-world data (cancer and dementia incidence, mortality), assuming no effect of cancer on dementia (true IRR = 1.00). To introduce competing risk of death, cancer history increased mortality. To introduce selective survival, we included a factor (prevalence ranging from 10% to 50%) that reduced cancer mortality and dementia incidence (IRRs ranged from 0.30 to 0.90). We calculated IRRs for cancer history on dementia incidence in the simulated cohorts. RESULTS Competing risk of death yielded unbiased cancer-dementia IRRs. With selective survival, bias was small (IRRs = 0.89 to 0.99), even under extreme scenarios. DISCUSSION The bias induced by selective survival in simulations was too small to explain the observed inverse cancer-dementia link, suggesting other mechanisms drive this association.
Collapse
Affiliation(s)
- Eleanor Hayes-Larson
- Department of Epidemiology, University of California, Los Angeles Fielding School of Public Health, Los Angeles, California, USA
| | - Sarah F Ackley
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
| | - Scott C Zimmerman
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
| | - Monica Ospina-Romero
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
| | - M Maria Glymour
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
| | - Rebecca E Graff
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
| | - John S Witte
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
| | - Lindsay C Kobayashi
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Elizabeth Rose Mayeda
- Department of Epidemiology, University of California, Los Angeles Fielding School of Public Health, Los Angeles, California, USA.,Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
| |
Collapse
|
194
|
McDade E, Bednar MM, Brashear HR, Miller DS, Maruff P, Randolph C, Ismail Z, Carrillo MC, Weber CJ, Bain LJ, Hake AM. The pathway to secondary prevention of Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2020; 6:e12069. [PMID: 32885024 PMCID: PMC7453146 DOI: 10.1002/trc2.12069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 07/09/2020] [Indexed: 11/11/2022]
Abstract
Alzheimer's disease (AD) is a continuum consisting of a preclinical stage that occurs decades before symptoms appear. As researchers make advances in investigating the continuum, the importance of developing drugs for secondary prevention is garnering increased discussion. For efficacious drug development for secondary prevention it is important to define what are the earliest biological stages of AD. The Alzheimer's Association Research Roundtable convened November 27 to 28, 2018 to focus on pre-clinical AD. This review will address the biological approach to defining pre-clinical AD, detection, identification of at-risk individuals, and lessons learned from trials such as A4 and TOMMORROW.
Collapse
Affiliation(s)
- Eric McDade
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Martin M. Bednar
- Takeda Pharmaceuticals International Co.Americas, Inc.CambridgeMassachusettsUSA
| | | | | | | | - Christopher Randolph
- MedAvante‐ProPhaseHamiltonNew JerseyUSA
- Department of NeurologyLoyola University Medical CenterMaywoodIllinoisUSA
| | - Zahinoor Ismail
- Cumming School of MedicineThe University of CalgaryCalgaryCanada
| | | | | | - Lisa J. Bain
- Independent Science WriterElversonPennsylvaniaUSA
| | - Ann Marie Hake
- Eli Lilly and CompanyIndianapolisIndianaUSA
- Department of NeurologyIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
195
|
Ternes K, Iyengar V, Lavretsky H, Dawson WD, Booi L, Ibanez A, Vahia I, Reynolds C, DeKosky S, Cummings J, Miller B, Perissinotto C, Kaye J, Eyre HA. Brain health INnovation Diplomacy: a model binding diverse disciplines to manage the promise and perils of technological innovation. Int Psychogeriatr 2020; 32:955-979. [PMID: 32019621 PMCID: PMC7423685 DOI: 10.1017/s1041610219002266] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Brain health diplomacy aims to influence the global policy environment for brain health (i.e. dementia, depression, and other mind/brain disorders) and bridges the disciplines of global brain health, international affairs, management, law, and economics. Determinants of brain health include educational attainment, diet, access to health care, physical activity, social support, and environmental exposures, as well as chronic brain disorders and treatment. Global challenges associated with these determinants include large-scale conflicts and consequent mass migration, chemical contaminants, air quality, socioeconomic status, climate change, and global population aging. Given the rapidly advancing technological innovations impacting brain health, it is paramount to optimize the benefits and mitigate the drawbacks of such technologies. OBJECTIVE We propose a working model of Brain health INnovation Diplomacy (BIND). METHODS We prepared a selective review using literature searches of studies pertaining to brain health technological innovation and diplomacy. RESULTS BIND aims to improve global brain health outcomes by leveraging technological innovation, entrepreneurship, and innovation diplomacy. It acknowledges the key role that technology, entrepreneurship, and digitization play and will increasingly play in the future of brain health for individuals and societies alike. It strengthens the positive role of novel solutions, recognizes and works to manage both real and potential risks of digital platforms. It is recognition of the political, ethical, cultural, and economic influences that brain health technological innovation and entrepreneurship can have. CONCLUSIONS By creating a framework for BIND, we can use this to ensure a systematic model for the use of technology to optimize brain health.
Collapse
Affiliation(s)
- Kylie Ternes
- School of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Vijeth Iyengar
- U.S. Administration on Aging/Administration for Community Living, U.S. Department of Health and Human Services, Washington, DC, USA
| | - Helen Lavretsky
- Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, UCLA, Los Angeles, California, USA
| | - Walter D Dawson
- Memory and Aging Center, School of Medicine, UCSF, San Francisco, California, USA
- Global Brain Health Institute, San Francisco, California, USA
- Trinity College Dublin, Dublin, Ireland
- School of Medicine, Oregon Health and Science University, Portland, Oregon, USA
- Institute on Aging, School of Urban and Public Affairs, Portland State University, Portland, Oregon, USA
| | - Laura Booi
- Global Brain Health Institute, San Francisco, California, USA
- Trinity College Dublin, Dublin, Ireland
| | - Agustin Ibanez
- Memory and Aging Center, School of Medicine, UCSF, San Francisco, California, USA
- Global Brain Health Institute, San Francisco, California, USA
- Trinity College Dublin, Dublin, Ireland
- Institute of Cognitive and Translational Neuroscience (INCYT), INECO Foundation, Favaloro University, Buenos Aires, Argentina
- National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
- Center for Social and Cognitive Neuroscience (CSCN), Universidad Adolfo Ibanez, Santiago, Chile
- Universidad Autónoma del Caribe, Barranquilla, Colombia
- ARC Centre of Excellence in Cognition and its Disorders, Sydney, Australia
| | - Ipsit Vahia
- McLean Hospital, Belmont, Massachusetts, USA
- Harvard Medical School, Cambridge, Massachusetts, USA
| | - Charles Reynolds
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Steven DeKosky
- McKnight Brain Institute and Department of Neurology, College of Medicine, University of Florida, Miami, Florida, USA
| | - Jeffrey Cummings
- Department of Brain Health, School of Integrated Health Sciences, Cleveland Clinic Lou Ruvo Center for Brain Health, UNLV, Las Vegas, Nevada, USA
| | - Bruce Miller
- Memory and Aging Center, School of Medicine, UCSF, San Francisco, California, USA
- Global Brain Health Institute, San Francisco, California, USA
- Trinity College Dublin, Dublin, Ireland
| | - Carla Perissinotto
- Division of Geriatrics, School of Medicine, UCSF, San Francisco, California, USA
| | - Jeffrey Kaye
- School of Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Harris A Eyre
- Innovation Institute, Texas Medical Center, Houston, Texas, USA
- Department of Psychiatry, University of Melbourne, Melbourne, Victoria, Australia
- IMPACT SRC, School of Medicine, Deakin University, Geelong, Victoria, Australia
- Brainstorm Laboratory for Mental Health Innovation, Department of Psychiatry, Stanford University School of Medicine, Palo Alto, California, USA
- Discipline of Psychiatry, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
196
|
Brain regions vulnerable and resistant to aging without Alzheimer's disease. PLoS One 2020; 15:e0234255. [PMID: 32726311 PMCID: PMC7390259 DOI: 10.1371/journal.pone.0234255] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 05/21/2020] [Indexed: 11/19/2022] Open
Abstract
'Normal aging' in the brain refers to age-related changes that occur independent of disease, in particular Alzheimer's disease. A major barrier to mapping normal brain aging has been the difficulty in excluding the earliest preclinical stages of Alzheimer's disease. Here, before addressing this issue we first imaged a mouse model and learn that the best MRI measure of dendritic spine loss, a known pathophysiological driver of normal aging, is one that relies on the combined use of functional and structural MRI. In the primary study, we then deployed the combined functional-structural MRI measure to investigate over 100 cognitively-normal people from 20-72 years of age. Next, to cover the tail end of aging, in secondary analyses we investigated structural MRI acquired from cognitively-normal people, 60-84 years of age, who were Alzheimer's-free via biomarkers. Collectively, the results from the primary functional-structural study, and the secondary structural studies revealed that the dentate gyrus is a hippocampal region differentially affected by aging, and that the entorhinal cortex is a region most resistant to aging. Across the cortex, the primary functional-structural study revealed and that the inferior frontal gyrus is differentially affected by aging, however, the secondary structural studies implicated other frontal cortex regions. Together, the results clarify how normal aging may affect the brain and has possible mechanistic and therapeutic implications.
Collapse
|
197
|
Canevelli M, Remoli G, Bacigalupo I, Valletta M, Toccaceli Blasi M, Sciancalepore F, Bruno G, Cesari M, Vanacore N. Use of Biomarkers in Ongoing Research Protocols on Alzheimer's Disease. J Pers Med 2020; 10:jpm10030068. [PMID: 32722106 PMCID: PMC7564515 DOI: 10.3390/jpm10030068] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023] Open
Abstract
The present study aimed to describe and discuss the state of the art of biomarker use in ongoing Alzheimer’s disease (AD) research. A review of 222 ongoing phase 1, 2, 3, and 4 protocols registered in the clinicaltrials.gov database was performed. All the trials (i) enrolling subjects with clinical disturbances and/or preclinical diagnoses falling within the AD continuum; and (ii) testing the efficacy and/or safety/tolerability of a therapeutic intervention, were analyzed. The use of biomarkers of amyloid deposition, tau pathology, and neurodegeneration among the eligibility criteria and/or study outcomes was assessed. Overall, 58.2% of ongoing interventional studies on AD adopt candidate biomarkers. They are mostly adopted by studies at the preliminary stages of the drug development process to explore the safety profile of novel therapies, and to provide evidence of target engagement and disease-modifying properties. The biologically supported selection of participants is mostly based on biomarkers of amyloid deposition, whereas the use of biomarkers as study outcomes mostly relies on markers of neurodegeneration. Biomarkers play an important role in the design and conduction of research protocols targeting AD. Nevertheless, their clinical validity, utility, and cost-effectiveness in the “real world” remain to be clarified.
Collapse
Affiliation(s)
- Marco Canevelli
- Department of Human Neuroscience, Sapienza University, 00185 Rome, Italy; (G.R.); (M.V.); (M.T.B.); (F.S.); (G.B.)
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, 00161 Rome, Italy; (I.B.); (N.V.)
- Correspondence: ; Tel./Fax: +39-(06)-4991-4604
| | - Giulia Remoli
- Department of Human Neuroscience, Sapienza University, 00185 Rome, Italy; (G.R.); (M.V.); (M.T.B.); (F.S.); (G.B.)
| | - Ilaria Bacigalupo
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, 00161 Rome, Italy; (I.B.); (N.V.)
| | - Martina Valletta
- Department of Human Neuroscience, Sapienza University, 00185 Rome, Italy; (G.R.); (M.V.); (M.T.B.); (F.S.); (G.B.)
| | - Marco Toccaceli Blasi
- Department of Human Neuroscience, Sapienza University, 00185 Rome, Italy; (G.R.); (M.V.); (M.T.B.); (F.S.); (G.B.)
| | - Francesco Sciancalepore
- Department of Human Neuroscience, Sapienza University, 00185 Rome, Italy; (G.R.); (M.V.); (M.T.B.); (F.S.); (G.B.)
| | - Giuseppe Bruno
- Department of Human Neuroscience, Sapienza University, 00185 Rome, Italy; (G.R.); (M.V.); (M.T.B.); (F.S.); (G.B.)
| | - Matteo Cesari
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy;
- Geriatric Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Nicola Vanacore
- National Center for Disease Prevention and Health Promotion, Italian National Institute of Health, 00161 Rome, Italy; (I.B.); (N.V.)
| |
Collapse
|
198
|
Diep C, Ftouni S, Manousakis JE, Nicholas CL, Drummond SPA, Anderson C. Acoustic slow wave sleep enhancement via a novel, automated device improves executive function in middle-aged men. Sleep 2020; 43:5613713. [PMID: 31691831 DOI: 10.1093/sleep/zsz197] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 05/21/2019] [Indexed: 02/06/2023] Open
Abstract
STUDY OBJECTIVES As slow-wave activity (SWA) is critical for cognition, SWA-enhancing technologies provide an exciting opportunity to improve cognitive function. We focus on improving cognitive function beyond sleep-dependent memory consolidation, using an automated device, and in middle-aged adults, who have depleted SWA yet a critical need for maximal cognitive capacity in work environments. METHODS Twenty-four healthy adult males aged 35-48 years participated in a randomized, double-blind, cross-over study. Participants wore an automated acoustic stimulation device that monitored real-time sleep EEG. Following an adaptation night, participants were exposed to either acoustic tones delivered on the up phase of the slow-wave (STIM) or inaudible "tones" during equivalent periods of stimulation (SHAM). An executive function test battery was administered after the experimental night. RESULTS STIM resulted in an increase in delta (0.5-4 Hz) activity across the full-night spectra, with enhancement being maximal at 1 Hz. SWA was higher for STIM relative to SHAM. Although no group differences were observed in any cognitive outcomes, due to large individual differences in SWA enhancement, higher SWA responders showed significantly improved verbal fluency and working memory compared with nonresponders. Significant positive associations were found between SWA enhancement and improvement in these executive function outcomes. CONCLUSIONS Our study suggests that (1) an automated acoustic device enhances SWA; (2) SWA enhancement improves executive function; (3) SWA enhancement in middle-aged men may be an important therapeutic target for enhancing cognitive function; and (4) there is a need to examine interindividual responses to acoustic stimulation and its effect on subsequent cognitive function. CLINICAL TRIAL REGISTRATION This study has been registered with the Australian New Zealand Clinical Trials Registry. "The efficacy of acoustic tones in slow-wave sleep enhancement and cognitive function in healthy adult males". https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=371548&isReview=true. REGISTRATION ACTRN12617000399392.
Collapse
Affiliation(s)
- Charmaine Diep
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Victoria, Australia.,Cooperative Research Centre for Alertness, Safety and Productivity, Notting Hill, Victoria, Australia
| | - Suzanne Ftouni
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Victoria, Australia.,Cooperative Research Centre for Alertness, Safety and Productivity, Notting Hill, Victoria, Australia
| | - Jessica E Manousakis
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Victoria, Australia
| | - Christian L Nicholas
- Melbourne School of Psychological Sciences, University of Melbourne, Melbourne, Victoria, Australia.,Institute for Breathing and Sleep, Austin Hospital, Melbourne, Victoria, Australia
| | - Sean P A Drummond
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Victoria, Australia
| | - Clare Anderson
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Victoria, Australia.,Cooperative Research Centre for Alertness, Safety and Productivity, Notting Hill, Victoria, Australia
| |
Collapse
|
199
|
Neuropsychological Test Performance and MRI Markers of Dementia Risk: Reducing Education Bias. Alzheimer Dis Assoc Disord 2020; 33:179-185. [PMID: 31206372 DOI: 10.1097/wad.0000000000000321] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND To use neuropsychological assessments for studying the underlying disease processes contributing to dementia, it is crucial that they correspond to magnetic resonance imaging (MRI)-based measures of dementia, regardless of educational level. METHODS French 3-City Dijon MRI study cohort members (n=1782) with assessments of white matter lesion volume (WMLV), hippocampal volume (HCV), and cerebrospinal fluid volume (CSFV), and 6 waves of neuropsychological assessments over 11 years, including Mini-Mental State Examination (MMSE), plus 5 other tests combined using a Z-score or item-response theory (IRT-cognition) comprised the study cohort. We evaluated, testing interactions, whether education modified associations of MRI markers with intercept or rate of change of MMSE, Z-score composite, or IRT-cognition. RESULTS In linear models, education modified the associations of WMLV and CSFV with MMSE and CSFV and Z-score composite. In mixed models, education modified the associations of WMLV and CSFV with level of MMSE and the association of HCV with slope of MMSE. Education also modified the association with CSFV and slope of Z-score composite decline. There was no evidence that education modified associations between MRI measures and level or slope of IRT-cognition. CONCLUSIONS Longitudinal analysis of correctly scaled neuropsychological assessments may provide unbiased proxies for MRI-based measures of dementia risk.
Collapse
|
200
|
Genistein and Galantamine Combinations Decrease β-Amyloid Peptide (1-42)-Induced Genotoxicity and Cell Death in SH-SY5Y Cell Line: an In Vitro and In Silico Approach for Mimic of Alzheimer's Disease. Neurotox Res 2020; 38:691-706. [PMID: 32613603 DOI: 10.1007/s12640-020-00243-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/09/2020] [Accepted: 06/12/2020] [Indexed: 01/09/2023]
Abstract
Alzheimer's disease (AD) is the primary dementia-causing disease worldwide, involving a multifactorial combination of environmental, genetic, and epigenetic factors, with essential participation of age and sex. Biochemically, AD is characterized by the presence of abnormal deposition of beta amyloid peptide (Aβ(1-42)), which in the brain is strongly correlated with oxidative stress, inflammation, DNA damage, and cholinergic impairment. The multiple mechanisms involved in its etiology create significant difficulty in producing an effective treatment. Neuroprotective properties of genistein and galantamine have been widely demonstrated through different mechanisms; however, it is unknown a possible synergistic neuroprotective effect against Aβ(1-42). In order to understand how genistein and galantamine combinations regulate the mechanisms of neuroprotection, we conducted a set of bioassays in vitro to evaluate cell viability, clonogenic survival, cell death, and anti-genotoxicity. Through molecular docking and therapeutic viability assays, we analyzed the inhibitory activity exerted by genistein on three major protein targets (AChE, BChE, and NMDA) involved in AD. The results showed that genistein and galantamine afforded significant protection at higher concentrations; however, combinations of sub-effective concentrations of both compounds provided marked neuroprotection when they were combined. In silico approaches showed that genistein has higher scores than the positive controls and low toxicity levels; nevertheless, the therapeutic viability indicated that unlike galantamine, genistein cannot undergo the action by P glycoprotein (PGP) and probably may be unable to cross the blood-brain barrier. In conclusion, our results show that genistein and galantamine exert neuroprotective by decreasing genotoxicity and cell death. In silico analysis, suggest that genistein modulates positively the expression of AChE, BChE, and NMDA. In this context, a combination of two or more drugs could inspire an attractive therapeutic strategy.
Collapse
|