201
|
Rosselli M, Tappen RM, Newman D. Semantic Interference Test: Evidence for Culture and Education Fairness from an Ethnically Diverse Sample in the USA. Arch Clin Neuropsychol 2019; 34:337-349. [PMID: 29688251 DOI: 10.1093/arclin/acy037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 03/01/2018] [Accepted: 04/12/2018] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE Tests of cognitive abilities are particularly susceptible to culture-based bias because these abilities are culturally bound. The specific purpose of this study was to examine the Semantic Interference Test, a clinical neuropsychological test, for culture bias. METHOD The sample included 415 community-dwelling participants (mean age 74, SD = 8.32; 308 were females) living in South Florida (USA). The sample included 72 African Americans, 93 Afro-Caribbeans, 77 Hispanic Americans, and 173 European Americans. An Item Response Theory analysis of bias was employed using the Differential Item Functioning (DIF) procedure. RESULTS Overall, the items appear to be invariant across gender, ethnicity, and education levels. Although the DIF identified several items that appear to differ across the two latter groupings, the multiple group confirmatory factor analysis (MG-CFA) suggests that these items have low impact on the overall measure. There were however, meaningful differences across age groups in the MG-CFA, suggesting that an age adjustment might be required. CONCLUSION The SIT can be considered a cognitive test that is not significantly affected by the participants' cultural background, at least within the culture range included in this study.
Collapse
Affiliation(s)
- Mónica Rosselli
- Department of Psychology, Charles E. Schmidt College of Science, Florida Atlantic University, Davie, FL, USA
| | - Ruth M Tappen
- Christine E. Lynn College of Nursing, Florida Atlantic University, Boca Raton, FL, USA
| | - David Newman
- Christine E. Lynn College of Nursing, Florida Atlantic University, Boca Raton, FL, USA
| |
Collapse
|
202
|
Langbaum JB, Karlawish J, Roberts JS, Wood EM, Bradbury A, High N, Walsh TL, Gordon D, Aggarwal R, Davis P, Stowell C, Trisko L, Langlois CM, Reiman EM, Tariot PN. GeneMatch: A novel recruitment registry using at-home APOE genotyping to enhance referrals to Alzheimer's prevention studies. Alzheimers Dement 2019; 15:515-524. [PMID: 30772251 PMCID: PMC6461487 DOI: 10.1016/j.jalz.2018.12.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/16/2018] [Accepted: 12/11/2018] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Recruitment for Alzheimer's disease (AD) prevention research studies is challenging because of lack of awareness among cognitively healthy adults coupled with the high screen fail rate due to participants not having a genetic risk factor or biomarker evidence of the disease. Participant recruitment registries offer one solution for efficiently and effectively identifying, characterizing, and connecting potential eligible volunteers to studies. METHODS Individuals aged 55-75 years who live in the United States and self-report not having a diagnosis of cognitive impairment such as MCI or dementia are eligible to join GeneMatch. Participants enroll online and are provided a cheek swab kit for DNA extraction and apolipoprotein E (APOE) genotyping. Participants are not told their APOE results, although the results may be used in part to help match participants to AD prevention studies. RESULTS As of August 2018, 75,351 participants had joined GeneMatch. Nearly 30% of participants have one APOE4 allele, and approximately 3% have two APOE4 alleles. The percentages of APOE4 heterozygotes and homozygotes are inversely associated with age (P < .001). DISCUSSION GeneMatch, the first trial-independent research enrollment program designed to recruit and refer cognitively healthy adults to AD prevention studies based in part on APOE test results, provides a novel mechanism to accelerate prescreening and enrollment for AD prevention trials.
Collapse
Affiliation(s)
| | - Jason Karlawish
- Department of Medical Ethics and Health Policy, University of Pennsylvania, Philadelphia, PA, USA
| | - J Scott Roberts
- Department of Health Behavior & Health Education, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Elisabeth M Wood
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Angela Bradbury
- Department of Medical Ethics and Health Policy, University of Pennsylvania, Philadelphia, PA, USA; Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nellie High
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| | | | | | | | | | | | | | | | - Eric M Reiman
- Banner Alzheimer's Institute, Phoenix, AZ, USA; Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, USA; Department of Psychiatry, University of Arizona, Tucson, AZ, USA; Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | | |
Collapse
|
203
|
|
204
|
Chen C, Anderson CS. Does reducing blood pressure and cholesterol provide any HOPE for preventing cognitive decline and dementia? Neurology 2019; 92:593-594. [DOI: 10.1212/wnl.0000000000007175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
205
|
Prato FS, Pavlosky WF, Foster SC, Thiessen JD, Beaujot RP. Screening for Dementia Caused by Modifiable Lifestyle Choices Using Hybrid PET/MRI. J Alzheimers Dis Rep 2019; 3:31-45. [PMID: 30842996 PMCID: PMC6400112 DOI: 10.3233/adr-180098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2018] [Indexed: 12/19/2022] Open
Abstract
Significant advances in positron emission tomography (PET) and magnetic resonance imaging (MRI) brain imaging in the early detection of dementia indicate that hybrid PET/MRI would be an effective tool to screen for dementia in the population living with lifestyle risk factors. Here we investigate the associated costs and benefits along with the needed imaging infrastructure. A demographic analysis determined the prevalence of dementia and its incidence. The expected value of the screening program was calculated assuming a sensitivity and specificity of 0.9, a prevalence of 0.1, a QALY factor of 0.348, a willingness to pay $114,000 CAD and the cost per PET/MRI scan of $2,000 CAD. It was assumed that each head PET/MRI could screen 3,000 individuals per year. The prevalence of dementia is increasing by almost two-fold every 20 years due to the increased population at ages where dementia is more prevalent. It has been shown that a five-year delay in the incidence of dementia would decrease the prevalence by some 45%. In Canada, a five-year delay corresponds to a health care savings of $27,000 CAD per subject per year. The expected value for screening was estimated at $23,745 CAD. The number of subjects to be screened per year in Canada, USA, and China between 60 and 79 was 11,405,000. The corresponding number of head-only hybrid PET/MRI systems needed is 3,800. A brain PET/MRI screening program is financially justifiable with respect to health care costs and justifies the continuing development of MRI compatible brain PET technology.
Collapse
Affiliation(s)
- Frank S. Prato
- Department of Medical Biophysics, Western University, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
- Department of Medical Imaging, Western University, London, ON, Canada
| | - William F. Pavlosky
- Lawson Health Research Institute, London, ON, Canada
- Department of Medical Imaging, Western University, London, ON, Canada
| | | | - Jonathan D. Thiessen
- Department of Medical Biophysics, Western University, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
- Department of Medical Imaging, Western University, London, ON, Canada
| | | |
Collapse
|
206
|
Wolters FJ, Tinga LM, Dhana K, Koudstaal PJ, Hofman A, Bos D, Franco OH, Ikram MA. Life Expectancy With and Without Dementia: A Population-Based Study of Dementia Burden and Preventive Potential. Am J Epidemiol 2019; 188:372-381. [PMID: 30299452 DOI: 10.1093/aje/kwy234] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 10/05/2018] [Indexed: 12/22/2022] Open
Abstract
Reliable population estimates of life expectancy with dementia are required for shaping health-care policy. From the Dutch, population-based Rotterdam Study, 10,348 persons were followed during 1990-2015 for dementia and death. We created multistate lifetables, and assessed the effect of postponing disease onset. During 120,673 person-years, 1,666 persons developed dementia, and 6,150 died. Overall life expectancy of women ranged from 18.0 years (95% confidence interval (CI): 17.8, 18.2) at age 65 to 2.3 years (95% CI: 2.2, 2.3) at age 95. Of total life expectancy at age 65, 5.7% (1.0 year (95% CI: 1.0, 1.1)) was lived with dementia, increasing with age to 42.1% (1.0 year, 95% CI: 0.9, 1.0) at age 95. For men, life expectancy ranged from 15.6 years (95% CI: 15.4, 15.9) at age 65 to 1.8 years (95% CI: 1.7, 1.8) at age 95, of which 3.7% (95% CI: 0.6 year, 0.5, 0.6) and 35.3% (95% CI: 0.6 year, 0.5, 0.7), respectively, was lived with dementia. Postponing dementia onset by 1-3 years resulted in 25%-57% reductions in years lived with dementia. Survival after diagnosis ranged from 6.7 years (95% CI: 5.3, 8.1) before age 70, to 2.6 years (95% CI: 2.3, 2.9) after age 90. The burden of dementia on individuals and society in terms of healthy life-years lost is large but could potentially be mitigated by preventive interventions at the population level.
Collapse
Affiliation(s)
- Frank J Wolters
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Liselotte M Tinga
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Klodian Dhana
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, United States of America
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | - Peter J Koudstaal
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Albert Hofman
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, United States of America
| | - Daniel Bos
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Oscar H Franco
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
207
|
Ding Y, Sohn JH, Kawczynski MG, Trivedi H, Harnish R, Jenkins NW, Lituiev D, Copeland TP, Aboian MS, Mari Aparici C, Behr SC, Flavell RR, Huang SY, Zalocusky KA, Nardo L, Seo Y, Hawkins RA, Hernandez Pampaloni M, Hadley D, Franc BL. A Deep Learning Model to Predict a Diagnosis of Alzheimer Disease by Using 18F-FDG PET of the Brain. Radiology 2019; 290:456-464. [PMID: 30398430 PMCID: PMC6358051 DOI: 10.1148/radiol.2018180958] [Citation(s) in RCA: 270] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/24/2018] [Accepted: 09/13/2018] [Indexed: 12/11/2022]
Abstract
Purpose To develop and validate a deep learning algorithm that predicts the final diagnosis of Alzheimer disease (AD), mild cognitive impairment, or neither at fluorine 18 (18F) fluorodeoxyglucose (FDG) PET of the brain and compare its performance to that of radiologic readers. Materials and Methods Prospective 18F-FDG PET brain images from the Alzheimer's Disease Neuroimaging Initiative (ADNI) (2109 imaging studies from 2005 to 2017, 1002 patients) and retrospective independent test set (40 imaging studies from 2006 to 2016, 40 patients) were collected. Final clinical diagnosis at follow-up was recorded. Convolutional neural network of InceptionV3 architecture was trained on 90% of ADNI data set and tested on the remaining 10%, as well as the independent test set, with performance compared to radiologic readers. Model was analyzed with sensitivity, specificity, receiver operating characteristic (ROC), saliency map, and t-distributed stochastic neighbor embedding. Results The algorithm achieved area under the ROC curve of 0.98 (95% confidence interval: 0.94, 1.00) when evaluated on predicting the final clinical diagnosis of AD in the independent test set (82% specificity at 100% sensitivity), an average of 75.8 months prior to the final diagnosis, which in ROC space outperformed reader performance (57% [four of seven] sensitivity, 91% [30 of 33] specificity; P < .05). Saliency map demonstrated attention to known areas of interest but with focus on the entire brain. Conclusion By using fluorine 18 fluorodeoxyglucose PET of the brain, a deep learning algorithm developed for early prediction of Alzheimer disease achieved 82% specificity at 100% sensitivity, an average of 75.8 months prior to the final diagnosis. © RSNA, 2018 Online supplemental material is available for this article. See also the editorial by Larvie in this issue.
Collapse
Affiliation(s)
- Yiming Ding
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Jae Ho Sohn
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Michael G. Kawczynski
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Hari Trivedi
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Roy Harnish
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Nathaniel W. Jenkins
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Dmytro Lituiev
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Timothy P. Copeland
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Mariam S. Aboian
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Carina Mari Aparici
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Spencer C. Behr
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Robert R. Flavell
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Shih-Ying Huang
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Kelly A. Zalocusky
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Lorenzo Nardo
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Youngho Seo
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Randall A. Hawkins
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Miguel Hernandez Pampaloni
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Dexter Hadley
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| | - Benjamin L. Franc
- From the Department of Radiology and Biomedical Imaging (Y.D.,
J.H.S., H.T., R.H., N.W.J., T.P.C., M.S.A., C.M.A., S.C.B., R.R.F., S.Y.H.,
Y.S., R.A.H., M.H.P., B.L.F.) and Institute for Computational Health Sciences
(J.H.S., M.G.K., H.T., D.L., K.A.Z., D.H.), University of California, San
Francisco, 550 Parnassus Ave, San Francisco, CA 94143; Department of Electrical
Engineering and Computer Sciences, University of California, Berkeley, Berkeley,
Calif (Y.D.); and Department of Radiology, University of California, Davis,
Sacramento, Calif (L.N.)
| |
Collapse
|
208
|
Abstract
PURPOSE OF REVIEW The aim of this review is to summarize current conceptual models of cognitive reserve (CR) and related concepts and to discuss evidence for these concepts within the context of aging and Alzheimer's disease. RECENT FINDINGS Evidence to date supports the notion that higher levels of CR, as measured by proxy variables reflective of lifetime experiences, are associated with better cognitive performance, and with a reduced risk of incident mild cognitive impairment/dementia. However, the impact of CR on longitudinal cognitive trajectories is unclear and may be influenced by a number of factors. Although there is promising evidence that some proxy measures of CR may influence structural brain measures, more research is needed. The protective effects of CR may provide an important mechanism for preserving cognitive function and cognitive well-being with age, in part because it can be enhanced throughout the lifespan. However, more research on the mechanisms by which CR is protective is needed.
Collapse
Affiliation(s)
- Corinne Pettigrew
- Department of Neurology, Johns Hopkins University School of Medicine, 1620 McElderry St., Reed Hall 1-West, Baltimore, MD, 21205, USA
| | - Anja Soldan
- Department of Neurology, Johns Hopkins University School of Medicine, 1620 McElderry St., Reed Hall 1-West, Baltimore, MD, 21205, USA.
| |
Collapse
|
209
|
Hahn C, Lee CU. A Brief Review of Paradigm Shifts in Prevention of Alzheimer's Disease: From Cognitive Reserve to Precision Medicine. Front Psychiatry 2019; 10:786. [PMID: 31736804 PMCID: PMC6837073 DOI: 10.3389/fpsyt.2019.00786] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 10/02/2019] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) and related dementias can be an enormous economic burden for taxpayers, patients, their families, medical systems, and society as a whole. Since disease-modifying treatments have failed, several studies have instead focused on a paradigm shift for preventing and treating AD. A higher cognitive reserve (e.g., greater education, occupational attainment, or more leisure activities) is associated with protection against disease-related cognitive decline. Precision medicine aims to optimize the effectiveness of disease prevention and treatment by considering specific biological components of individuals. We suggest that research into cognitive reserve and precision medicine could be a key to overcoming the limitations of traditional approaches to the prevention and treatment of AD.
Collapse
Affiliation(s)
- Changtae Hahn
- Department of Psychiatry, Deajeon Saint Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Chang Uk Lee
- Department of Psychiatry, Seoul Saint Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
210
|
Zimering MB, Patel D, Bahn G. Type 2 Diabetes Predicts Increased Risk of Neurodegenerative Complications in Veterans Suffering Traumatic Brain Injury. JOURNAL OF ENDOCRINOLOGY AND DIABETES 2019; 6:137. [PMID: 31828222 PMCID: PMC6905496 DOI: 10.15226/2374-6890/6/3/001137] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
AIMS Obese type 2 diabetes and traumatic brain injury are associated with persistent peripheral and neuro-inflammation, respectively. We tested whether adult type 2 diabetes increased the hazard rate for neurodegeneration complications following traumatic brain injury. METHODS Retrospective chart review of patients treated at the Veterans Affairs New Jersey Healthcare System between 2016-2019 and having a diagnosis of prior traumatic brain injury was performed in adult veterans, age 50 years or older. Cox proportional hazards regression analysis was used to identify risk factors predictive of an increased risk of neurodegeneration, i.e. worsening major depression, dementia or Parkinson's disease following traumatic brain injury. RESULTS Type 2 diabetes predicted a nearly three-fold increased hazard ratio (HR = 2.95, 95% CI 1.15-7.56, P =0.02) for the occurrence of worsening major depression, dementia or Parkinson's disease in eighty adults age 50 years or older who had experienced prior traumatic brain injury. After adjusting for other covariates, hypertension (HR= 4.15, 95% CI 1.21-14.29, P =0.02) was significant and body mass index (HR=1.14, 95% CI 0.99-1.30; P=0.06) modestly significant predictors of the risk for the time to first occurrence of the composite neurodegenerative outcome. CONCLUSION Type 2 diabetes, hypertension and higher body mass index increase the hazard for the occurrence of worsening depression, Parkinson's disease and dementia following traumatic brain injury in middle-aged and older adults.
Collapse
Affiliation(s)
- Mark B. Zimering
- Endocrinology, Veterans Affairs New Jersey Healthcare System, East Orange, NJ
- Rutgers-Robert Wood Johnson Medical School, New Brunswick NJ, USA
| | - Deesha Patel
- Endocrinology, Veterans Affairs New Jersey Healthcare System, East Orange, NJ
| | - Gideon Bahn
- Hines Veterans Affairs Hospital, Hines, Illinois
| |
Collapse
|
211
|
Yagi S, Galea LAM. Sex differences in hippocampal cognition and neurogenesis. Neuropsychopharmacology 2019; 44:200-213. [PMID: 30214058 PMCID: PMC6235970 DOI: 10.1038/s41386-018-0208-4] [Citation(s) in RCA: 217] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 08/29/2018] [Accepted: 08/30/2018] [Indexed: 12/27/2022]
Abstract
Sex differences are reported in hippocampal plasticity, cognition, and in a number of disorders that target the integrity of the hippocampus. For example, meta-analyses reveal that males outperform females on hippocampus-dependent tasks in rodents and in humans, furthermore women are more likely to experience greater cognitive decline in Alzheimer's disease and depression, both diseases characterized by hippocampal dysfunction. The hippocampus is a highly plastic structure, important for processing higher order information and is sensitive to the environmental factors such as stress. The structure retains the ability to produce new neurons and this process plays an important role in pattern separation, proactive interference, and cognitive flexibility. Intriguingly, there are prominent sex differences in the level of neurogenesis and the activation of new neurons in response to hippocampus-dependent cognitive tasks in rodents. However, sex differences in spatial performance can be nuanced as animal studies have demonstrated that there are task, and strategy choice dependent sex differences in performance, as well as sex differences in the subregions of the hippocampus influenced by learning. This review discusses sex differences in pattern separation, pattern completion, spatial learning, and links between adult neurogenesis and these cognitive functions of the hippocampus. We emphasize the importance of including both sexes when studying genomic, cellular, and structural mechanisms of the hippocampal function.
Collapse
Affiliation(s)
- Shunya Yagi
- Department of Psychology, Graduate Program in Neuroscience, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Liisa A M Galea
- Department of Psychology, Graduate Program in Neuroscience, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
212
|
Sur M, Dey P, Sarkar A, Bar S, Banerjee D, Bhat S, Mukherjee P. Sarm1 induction and accompanying inflammatory response mediates age-dependent susceptibility to rotenone-induced neurotoxicity. Cell Death Discov 2018; 4:114. [PMID: 30564462 PMCID: PMC6289984 DOI: 10.1038/s41420-018-0119-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/29/2018] [Accepted: 10/16/2018] [Indexed: 11/12/2022] Open
Abstract
Aging is a complex biological process and environmental risk factors like pesticide exposure have been implicated in the increased incidence of age-related neurodegenerative diseases like Parkinson’s disease (PD) but the etiology remains unknown. There is also lack of a proper animal model system to study the progressive effect of these environmental toxins on age-associated neurodegeneration. In this study, we established a drosophila model of aging to study the age-dependent vulnerability to the environmental toxin rotenone that has been implicated in sporadic cases of PD. We demonstrate that age plays a determining role in the increased susceptibility to chronic rotenone exposure that is accompanied by severe locomotor deficits, decreased lifespan and loss of dopaminergic (DA) neurons. Chronic low dose exposure to rotenone results in the rapid induction of the neurodegenerative molecule SARM1/dSarm. Further, the age-dependent dSarm induction is accompanied by a heightened inflammatory response (increased expression of Eiger and Relish) that is independent of reactive oxygen species (ROS) generation in the observed rotenone-induced neurotoxicity. dSarm induction and subsequent locomotor deficits is reversed in the presence of the anti-inflammatory molecule resveratrol. Thus, dSarm and heightened inflammatory responses may play a crucial role in age-dependent vulnerability to the pesticide rotenone thus making it an attractive target to help develop cost-effective therapeutic strategies to prevent ongoing dopaminergic neuronal loss as seen in PD.
Collapse
Affiliation(s)
- Malinki Sur
- 1Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata, 700073 West Bengal India
| | - Puja Dey
- 1Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata, 700073 West Bengal India
| | - Ankita Sarkar
- 1Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata, 700073 West Bengal India
| | - Sudipta Bar
- 2Department of Biological Sciences, IISER Kolkata, Mohanpur, 741246 West Bengal India
| | - Dipanjana Banerjee
- 1Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata, 700073 West Bengal India
| | - Swati Bhat
- 1Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata, 700073 West Bengal India
| | - Piyali Mukherjee
- 1Department of Life Sciences, Presidency University, 86/1, College Street, Kolkata, 700073 West Bengal India
| |
Collapse
|
213
|
Vable AM, Eng CW, Mayeda ER, Basu S, Marden JR, Hamad R, Glymour MM. Mother's education and late-life disparities in memory and dementia risk among US military veterans and non-veterans. J Epidemiol Community Health 2018; 72:1162-1167. [PMID: 30082424 PMCID: PMC6226315 DOI: 10.1136/jech-2018-210771] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/06/2018] [Accepted: 07/11/2018] [Indexed: 11/03/2022]
Abstract
BACKGROUND Adverse childhood socioeconomic status (cSES) predicts higher late-life risk of memory loss and dementia. Veterans of U.S. wars are eligible for educational and economic benefits that may offset cSES disadvantage. We test whether cSES disparities in late-life memory and dementia are smaller among veterans than non-veterans. METHODS Data came from US-born men in the 1995-2014 biennial surveys of the Health and Retirement Study (n=7916 born 1928-1956, contributing n=38 381 cognitive assessments). Childhood SES was represented by maternal education. Memory and dementia risk were assessed with brief neuropsychological assessments and proxy reports. Military service (veteran/non-veteran) was evaluated as a modifier of the effect of maternal education on memory and dementia risk. We employed linear or logistic regression models to test whether military service modified the effect of maternal education on memory or dementia risk, adjusted for age, race, birthplace and childhood health. RESULTS Low maternal education was associated with worse memory than high maternal education (β = -0.07 SD, 95% CI -0.08 to -0.05), while veterans had better memory than non-veterans (β = 0.03 SD, 95% CI 0.02 to 0.04). In interaction analyses, maternal education disparities in memory were smaller among veterans than non-veterans (difference in disparities = 0.04 SD, 95% CI 0.01 to 0.08, p = 0.006). Patterns were similar for dementia risk. CONCLUSIONS Disparities in memory by maternal education were smaller among veterans than non-veterans, suggesting military service and benefits partially offset the deleterious effects of low maternal education on late-life cognitive outcomes.
Collapse
Affiliation(s)
- Anusha M Vable
- Center for Population Health Sciences, Stanford University, Palo Alto, California, USA.,Center for Primary Care and Outcomes Research, Stanford University, Palo Alto, California, USA.,Department of Family and Community Medicine, University of California, San Francisco, San Francisco, California, USA.,Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, USA
| | - Chloe W Eng
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, USA
| | - Elizabeth Rose Mayeda
- Department of Epidemiology, University of California, Los Angeles, Los Angeles, California, USA
| | - Sanjay Basu
- Center for Population Health Sciences, Stanford University, Palo Alto, California, USA.,Center for Primary Care and Outcomes Research, Stanford University, Palo Alto, California, USA.,Center for Primary Care, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Rita Hamad
- Department of Family and Community Medicine, University of California, San Francisco, San Francisco, California, USA.,Philip R. Lee Institute for Health Policy Studies, University of California, San Francisco, San Francisco, California, USA
| | - M Maria Glymour
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, USA.,Department of Social and Behavioral Sciences, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
214
|
Bundy JL, Vied C, Badger C, Nowakowski RS. Sex-biased hippocampal pathology in the 5XFAD mouse model of Alzheimer's disease: A multi-omic analysis. J Comp Neurol 2018; 527:462-475. [PMID: 30291623 DOI: 10.1002/cne.24551] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 08/16/2018] [Accepted: 09/25/2018] [Indexed: 01/01/2023]
Abstract
Alzheimer's disease is a progressive neurodegenerative disorder and the most common form of dementia. Like many neurological disorders, Alzheimer's disease has a sex-biased epidemiological profile, affecting approximately twice as many women as men. The cause of this sex difference has yet to be elucidated. To identify molecular correlates of this sex bias, we investigated molecular pathology in females and males using the 5XFamilial Alzheimer's disease mutations (5XFAD) genetic mouse model of Alzheimer's disease. We profiled the transcriptome and proteome of the mouse hippocampus during early stages of disease development (1, 2, and 4 months of age). Our analysis reveals 42 genes that are differentially expressed between disease and wild-type animals at 2 months of age, prior to observable plaque deposition. In 4-month-old animals, we detect 1,316 differentially expressed transcripts between transgenic and control 5XFAD mice, many of which are associated with immune function. Additionally, we find that some of these transcriptional perturbations are correlated with altered protein levels in 4-month-old transgenic animals. Importantly, our data indicate that female 5XFAD mouse exhibit more profound pathology than their male counterparts as measured by differences in gene expression. We also find that the 5XFAD transgenes are more highly expressed in female 5XFAD mice than their male counterparts, which could partially account for the sex-biased molecular pathology observed in this dataset.
Collapse
Affiliation(s)
- Joseph L Bundy
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida
| | - Cynthia Vied
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida.,Translational Science Laboratory, Florida State University College of Medicine, Tallahassee, Florida
| | - Crystal Badger
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida
| | - Richard S Nowakowski
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida
| |
Collapse
|
215
|
Li Y, Yao Z, Zhang H, Hu B. Indirect relation based individual metabolic network for identification of mild cognitive impairment. J Neurosci Methods 2018; 309:188-198. [DOI: 10.1016/j.jneumeth.2018.09.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/05/2018] [Accepted: 09/03/2018] [Indexed: 11/16/2022]
|
216
|
Higher Mediterranean Diet scores are not cross-sectionally associated with better cognitive scores in 20- to 70-year-old Dutch adults: The NQplus study. Nutr Res 2018; 59:80-89. [DOI: 10.1016/j.nutres.2018.07.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 07/22/2018] [Accepted: 07/27/2018] [Indexed: 12/13/2022]
|
217
|
Design, synthesis and biological evaluation of 1,4-Diazobicylco[3.2.2]nonane derivatives as α7-Nicotinic acetylcholine receptor PET/CT imaging agents and agonists for Alzheimer's disease. Eur J Med Chem 2018; 159:255-266. [PMID: 30296684 DOI: 10.1016/j.ejmech.2018.09.064] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 09/20/2018] [Accepted: 09/25/2018] [Indexed: 11/20/2022]
Abstract
α7-Nicotinic acetylcholine receptor (α7-nAChR) agonists are promising therapeutic drug candidates for treating the cognitive impairment associated with Alzheimer's disease (AD). Thus, a novel class of derivatives of 1,4-diazobicylco[3.2.2]nonane has been synthesized and evaluated as α7-nAChR ligands. Five of them displayed high binding affinity (Ki = 0.001-25 nM). In particular, the Ki of 14 was 0.0069 nM, which is superior to that of the most potent ligand that was previously reported by an order of magnitude. Four of them had high selectivity for α7-nAChRs over α4β2-nAChRs and no significant hERG (human ether-a-go-go-related gene) inhibition. Their agonist activity was also discussed preliminarily. One of the compounds, 15 (Ki = 2.98 ± 1.41 nM), was further radiolabeled with 18F to afford [18F]15 for PET imaging, which exhibited high initial brain uptake (11.60 ± 0.14%ID/g at 15 min post injection), brain/blood value (9.57 at 30 min post injection), specific labeling of α7-nAChRs and fast clearance from the brain. Blocking studies demonstrated that [18F]15 was α7-nAChR selective. In addition, micro-PET/CT imaging in normal rats further indicated that [18F]15 had obvious accumulation in the brain. Therefore, [18F]15 was proved to be a potential PET radiotracer for α7-nAChR imaging.
Collapse
|
218
|
Banach M, Konieczny L, Wiśniowski Z, Roterman I. Fragment Aβ(18-41) presented within the CDR3 loop region of a shark immunoglobulin new antigen receptor single-variable domain antibody analyzed based on the fuzzy oil drop model. BIO-ALGORITHMS AND MED-SYSTEMS 2018. [DOI: 10.1515/bams-2018-0028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
The structure of amyloid Aβ(1-41) is the object of many papers due to the neurodegenerative processes induced by this amyloid. One of the ways to investigate the possible structural forms other than the amyloid is to incorporate the fragment of this peptide into the chain of immunoglobulin. Fragment Aβ(18-41) presented within the CDR3 loop region of a shark immunoglobulin new antigen receptor single-variable domain antibody is the object of this analysis. The structure of this hybrid is available in the PDB and analyzed based on the fuzzy oil drop model. The aim is to define the status of this fragment, revealing the possible fitting to the ordered form of the hydrophobic core. Simultaneously, the verification of the predisposition to complexation is possible.
Collapse
|
219
|
Espinosa Del Pozo PH, Espinosa PS, Donadi EA, Martinez EZ, Salazar-Uribe JC, Guerrero MA, Moriguti JC, Colcha MC, Garcia SE, Naranjo R, Altamirano WE, Koek AY. Cognitive Decline in Adults Aged 65 and Older in Cumbayá, Quito, Ecuador: Prevalence and Risk Factors. Cureus 2018; 10:e3269. [PMID: 30430058 PMCID: PMC6221535 DOI: 10.7759/cureus.3269] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Objective To assess the prevalence of and risk factors for cognitive decline and dementia in individuals greater than 65 years of age in Cumbayá, Quito, Ecuador. Methods This is a cross-sectional observational study that was carried out in adults over age 65. The Mini Mental State Examination (MMSE), Ascertain Dementia Eight-Item Informant Questionnaire (AD8), and Mini Nutritional Assessment (MNA) were used to assess the cognitive status and nutritional habits of this population. Results A total of 144 patients (mean age 75.3 years, 77.1% female) participated in this study. Forty percent of patients had AD8 and MMSE scores consistent with cognitive impairment and possible dementia. Age (p < 0.01), lower educational level (p < 0.01), history of stroke (p < 0.01), history of intracerebral hemorrhage (p < 0.01), diabetes mellitus (p < 0.01), and malnutrition (p < 0.01) were statistically significant risk factors for cognitive impairment. Exercise was found to be protective against cognitive decline in our study group (p < 0.03). Gender, ethnicity, location, head trauma, Parkinson disease, hypercholesterolemia, myocardial infarction, thyroid disease, depression, anxiety, and family history of dementia were not found to be associated with cognitive decline in this population. Conclusions The prevalence of cognitive impairment and possible dementia is 18–21% at age 65 and 54–60% at age 85 in Cumbayá, Quito, Ecuador. The major risk factors for cognitive impairment in this population are age, low educational level, malnutrition, prior stroke, prior intracerebral hemorrhage, and diabetes. Protective factors for cognitive decline include exercise and possibly modest consumption of alcohol.
Collapse
Affiliation(s)
| | - Patricio S Espinosa
- Neurology, Marcus Neuroscience Institute, Boca Raton Regional Hospital, Boca Raton, USA
| | - Eduardo A Donadi
- Departamento De Clinica, Facultad De Medicina Ribeirão Preto, Universidad De São Paulo, Ribeirão Preto, BRA
| | - Edson Z Martinez
- Social Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, BRA
| | - Juan C Salazar-Uribe
- Statistics, Universidad Nacional De Colombia (national University of Colombia), Medellín, COL
| | - Marco A Guerrero
- Neurosciences Unit, College of Medical Sciences, Central University of Ecuador, Quito, ECU
| | - Julio C Moriguti
- Departamento De Geriatria, Facultad De Medicina Ribeirão Preto, Universidad De São Paulo, Ribeirão, BRA
| | - Mishell C Colcha
- Neurosciences Unit, College of Medical Sciences, Central University of Ecuador, Quito, ECU
| | - Susana E Garcia
- Unidad De Neurociencias, Facultad De Ciencias Médicas, Universidad Central Del Ecuador, Quito, ECU
| | - Raquel Naranjo
- Neurosciences Unit, College of Medical Sciences, Central University of Ecuador, Quito, ECU
| | - Wilson E Altamirano
- Neurosciences Unit, College of Medical Sciences, Central University of Ecuador, Quito, ECU
| | - Adriana Y Koek
- Clinical Biomedical Science, Florida Atlantic University Charles E. Schmidt College of Medicine, Boca Raton, USA
| |
Collapse
|
220
|
Irvine MA, Scholey A, King R, Gillings R, Vauzour D, Demichele SJ, Das T, Wesnes KA, Sutton BP, Cassidy A, Pipingas A, Potter JF, Johnson G, White D, Larsen R, Cohen NJ, Minihane AM. The Cognitive Ageing, Nutrition and Neurogenesis (CANN) trial: Design and progress. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2018; 4:591-601. [PMID: 30426067 PMCID: PMC6222033 DOI: 10.1016/j.trci.2018.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Introduction The Cognitive Ageing, Nutrition and Neurogenesis trial hypothesizes that a combined intervention with long-chain n-3 polyunsaturated fatty acids (n-3) and cocoa flavan-3-ols (FLAV) will mitigate the cognitive decline anticipated to naturally occur over 1 year in older adults. Methods In a double-blinded, placebo-controlled parallel design, 259 individuals with mild cognitive impairment or subjective memory impairment were randomized to a control or n-3 FLAV group (1.5 g docosahexaenoic acid + eicosapentaenoic acid and 500 mg n-3 FLAV daily) for 12 months. Cognition was measured at 0, 3, and 12 months. The primary end-point is hippocampus-sensitive cognitive function (e.g., number of false-positives on the Picture Recognition Task of the Cognitive Drug Research test battery). Secondary outcomes include additional cognitive measures, brain atrophy and blood flow (assessed by magnetic resonance imaging), vascular function, circulating biomarkers of cardiovascular and cognitive health, gut microflora, red blood cell fatty acid status, and urine flavan-3-ol metabolites. Results Screening began in 2015, with all baseline visits completed in March 2017. The intervention was finished in March 2018. Discussion Cognitive Ageing, Nutrition and Neurogenesis aims to identify an effective diet-based intervention to prevent or delay cognitive impairment in cognitively at-risk individuals, which could ultimately contribute to a reduced population burden of dementia. Clinicaltrials.gov NCT02525198.
Collapse
Affiliation(s)
- Michael A Irvine
- Department of Nutrition and Preventive Medicine, Norwich Medical School, BCRE, University of East Anglia, Norwich, UK
| | - Andrew Scholey
- Centre for Human Psychopharmacology, Swinburne University of Technology, Hawthorn, VIC, Australia
| | - Rebecca King
- Centre for Human Psychopharmacology, Swinburne University of Technology, Hawthorn, VIC, Australia
| | - Rachel Gillings
- Department of Nutrition and Preventive Medicine, Norwich Medical School, BCRE, University of East Anglia, Norwich, UK
| | - David Vauzour
- Department of Nutrition and Preventive Medicine, Norwich Medical School, BCRE, University of East Anglia, Norwich, UK
| | - Stephen J Demichele
- Strategic Research and Development, Abbott Nutrition, Abbott Laboratories, Columbus, OH, USA
| | - Tapas Das
- Strategic Research and Development, Abbott Nutrition, Abbott Laboratories, Columbus, OH, USA
| | - Keith A Wesnes
- University of Exeter Medical School, St Luke's Campus, Exeter, UK
| | - Brad P Sutton
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbane, IL, USA.,Centre for Nutrition Learning and Memory, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbane, IL, USA
| | - Aedin Cassidy
- Department of Nutrition and Preventive Medicine, Norwich Medical School, BCRE, University of East Anglia, Norwich, UK
| | - Andrew Pipingas
- Centre for Human Psychopharmacology, Swinburne University of Technology, Hawthorn, VIC, Australia
| | - John F Potter
- Department of Nutrition and Preventive Medicine, Norwich Medical School, BCRE, University of East Anglia, Norwich, UK
| | - Glyn Johnson
- Department of Nutrition and Preventive Medicine, Norwich Medical School, BCRE, University of East Anglia, Norwich, UK
| | - David White
- Centre for Human Psychopharmacology, Swinburne University of Technology, Hawthorn, VIC, Australia
| | - Ryan Larsen
- Strategic Research and Development, Abbott Nutrition, Abbott Laboratories, Columbus, OH, USA
| | - Neal J Cohen
- Strategic Research and Development, Abbott Nutrition, Abbott Laboratories, Columbus, OH, USA.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbane, IL, USA
| | - Anne-Marie Minihane
- Department of Nutrition and Preventive Medicine, Norwich Medical School, BCRE, University of East Anglia, Norwich, UK
| |
Collapse
|
221
|
The effect of APOE genotype on Alzheimer's disease risk is influenced by sex and docosahexaenoic acid status. Neurobiol Aging 2018; 69:209-220. [DOI: 10.1016/j.neurobiolaging.2018.05.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 04/23/2018] [Accepted: 05/14/2018] [Indexed: 01/21/2023]
|
222
|
Haines JL. Alzheimer Disease: Perspectives from Epidemiology and Genetics. THE JOURNAL OF LAW, MEDICINE & ETHICS : A JOURNAL OF THE AMERICAN SOCIETY OF LAW, MEDICINE & ETHICS 2018; 46:694-698. [PMID: 30336113 DOI: 10.1177/1073110518804230] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Alzheimer disease (AD) is a huge and growing societal problem with upwards of 35% of the population over the age of 80 developing the disease. AD results in a loss of memory, the ability to make reasoned and sound decisions, and ultimately the inability to take care of oneself. AD has an impact not only on the sufferer, but their caretakers and loved ones, who must take on a costly and time-consuming burden of care. AD is found in virtually all racial and ethnic groups. Genetic influences on AD are substantial, and there has been a 30 year history of both success and failure. Mutations for rare early onset forms of the disease have been identified, but this information has not yet led to an effective treatment. Multiple common genetic variations have also been identified, and have led to new insights into the potential role of microglia cells in addition to neuronal cells in the brain. Despite intensive efforts, a significant portion of the genetic etiology of AD remains unknown and must be identified.
Collapse
Affiliation(s)
- Jonathan L Haines
- Jonathan L. Haines, Ph.D., is Professor and Chair in the Department of Population & Quantitative Health Sciences at Case Western Reserve School of Medicine. He received his B.A. from Colby College (Waterville, ME) and his Ph.D. in Genetics and Cell Biology from the University of Minnesota (Minneapolis)
| |
Collapse
|
223
|
Bott N, Kumar S, Krebs C, Glenn JM, Madero EN, Juusola JL. A Remote Intervention to Prevent or Delay Cognitive Impairment in Older Adults: Design, Recruitment, and Baseline Characteristics of the Virtual Cognitive Health (VC Health) Study. JMIR Res Protoc 2018; 7:e11368. [PMID: 30104186 PMCID: PMC6111147 DOI: 10.2196/11368] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 07/23/2018] [Indexed: 12/31/2022] Open
Abstract
Background A growing body of evidence supports the use of lifestyle interventions for preventing or delaying the onset of Alzheimer disease and other forms of dementia in at-risk individuals. The development of internet-delivered programs would increase the scalability and reach of these interventions, but requires validation to ensure similar effectiveness to brick-and-mortar options. Objective We describe the study design, recruitment process, and baseline participant characteristics of the sample in the Virtual Cognitive Health (VC Health) study. Future analyses will assess the impact of the remotely delivered lifestyle intervention on (1) cognitive function, (2) depression and anxiety, and (3) various lifestyle behaviors, including diet, exercise, and sleep, in a cohort of older adults with subjective memory decline. Additional analyses will explore feasibility outcomes, as well as the participants’ engagement patterns with the program. Methods Older adults (aged 60-75 years) with subjective memory decline as measured by the Subjective Cognitive Decline 9-item (SCD-9) questionnaire, and who reported feeling worried about their memory decline, were eligible to participate in this single-arm pre-post study. All participants enrolled in the yearlong digital intervention, which consists of health coach-guided lifestyle change for improving diet, exercise, sleep, stress, and cognition. All components of this study were conducted remotely, including the collection of data and the administration of the intervention. We assessed participants at baseline, 12 weeks, 24 weeks, and 52 weeks with online surveys and the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) test. We will conduct intention-to-treat analysis on all outcomes. Results A total of 85 participants enrolled in the intervention and 82 are included in the study sample (3 participants withdrew). The study cohort of 82 participants comprises 61 (74%) female, 72 (88%) white, and 64 (78%) overweight or obese participants, and 55 (67%) have at least a college degree. The average baseline RBANS score was 95.9 (SD 11.1), which is within age-adjusted norms. The average SCD-9 score was 6.0 (SD 2.0), indicating minor subjective cognitive impairment at the beginning of the study. The average baseline Generalized Anxiety Disorder 7-item scale score was 6.2 (SD 4.5), and the average Patient Health Questionnaire 9-item score was 8.5 (SD 4.9), indicating mild levels of anxiety and depression at baseline. Conclusions Internet-delivered lifestyle interventions are a scalable solution for the prevention or delay of Alzheimer disease. The results of this study will provide the first evidence for the effectiveness of a fully remote intervention and lay the groundwork for future investigations. Trial Registration ClinicalTrials.gov NCT02969460; http://clinicaltrials.gov/ct2/show/NCT02969460 (Archived by WebCite at http://www.webcitation.org/71LkYAkSh) Registered Report Identifier RR1-10.2196/11368
Collapse
Affiliation(s)
- Nicholas Bott
- Department of Medicine, School of Medicine, Stanford University, Stanford, CA, United States.,Neurotrack Technologies, Inc, Redwood City, CA, United States
| | | | - Caitlyn Krebs
- Neurotrack Technologies, Inc, Redwood City, CA, United States
| | - Jordan M Glenn
- Neurotrack Technologies, Inc, Redwood City, CA, United States
| | - Erica N Madero
- Neurotrack Technologies, Inc, Redwood City, CA, United States
| | | |
Collapse
|
224
|
Noreen Z, DeJesus J, Bhatti A, Loffredo CA, John P, Khan JS, Nunlee-Bland G, Ghosh S. Epidemiological Investigation of Type 2 Diabetes and Alzheimer's Disease in a Pakistani Population. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:E1582. [PMID: 30049934 PMCID: PMC6122092 DOI: 10.3390/ijerph15081582] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/19/2018] [Accepted: 07/23/2018] [Indexed: 01/01/2023]
Abstract
The epidemic of type 2 diabetes mellitus (T2DM) and the possibility of it contributing to the risk of Alzheimer's disease (AD) have become important health concerns worldwide and in Pakistan, where the co-occurrence of T2DM and AD is becoming more frequent. To gain insights on this phenomenon, a cross-sectional study was initiated. We recruited and interviewed 820 research participants from four cities in Pakistan: 250 controls, 450 T2DM, 100 AD, and 20 with both diseases. Significant differences between groups were observed for age (p < 0.0001), urban vs. rural locality (p = 0.0472) and residing near industrial areas. The average HbA1c (%) level was 10.68 ± 2.34 in the T2DM group, and females had a lower level than males (p = 0.003). In the AD group, significant relationships existed between education and family history. Overall, the results suggest that T2DM and AD were associated with both socio-demographic and environmental factors in Pakistani participants. Detailed molecular investigations are underway in our laboratory to decipher the differential genetic pathways of the two diseases to address their increasing prevalence in this developing nation.
Collapse
Affiliation(s)
- Zarish Noreen
- Department of Biology, Howard University, Washington, DC 20059, USA.
- Department of Healthcare Biotechnology, National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan.
| | - Jessica DeJesus
- Departments of Oncology and of Biostatistics, Georgetown University, Washington, DC 20057, USA.
| | - Attya Bhatti
- Department of Healthcare Biotechnology, National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan.
| | - Christopher A Loffredo
- Departments of Oncology and of Biostatistics, Georgetown University, Washington, DC 20057, USA.
| | - Peter John
- Department of Healthcare Biotechnology, National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan.
| | - Jahangir S Khan
- Department of Surgery, Rawalpindi Medical College, Rawalpindi, Punjab 46000, Pakistan.
| | - Gail Nunlee-Bland
- Departments of Pediatrics and Child Health, College of Medicine, Howard University, Washington, DC 20059, USA.
| | - Somiranjan Ghosh
- Department of Biology, Howard University, Washington, DC 20059, USA.
- Departments of Pediatrics and Child Health, College of Medicine, Howard University, Washington, DC 20059, USA.
| |
Collapse
|
225
|
Peripheral Biomarkers for Early Detection of Alzheimer's and Parkinson's Diseases. Mol Neurobiol 2018; 56:2256-2277. [PMID: 30008073 DOI: 10.1007/s12035-018-1151-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 05/24/2018] [Indexed: 01/18/2023]
Abstract
Neurological disorders are found to be influencing the peripheral tissues outside CNS. Recent developments in biomarkers for CNS have emerged with various diagnostic and therapeutic shortcomings. The role of central biomarkers including CSF-based and molecular imaging-based probes are still unclear for early diagnosis of major neurological diseases. Current trends show that early detection of neurodegenerative diseases with non-invasive methods is a major focus of researchers, and the development of biomarkers aiming peripheral tissues is in demand. Alzheimer's and Parkinson's diseases are known for the progressive loss in neural structures or functions, including the neural death. Various dysfunctions of metabolic and biochemical pathways are associated with early occurrence of neuro-disorders in peripheral tissues including skin, blood cells, and eyes. This article reviews the peripheral biomarkers explored for early detection of Alzheimer's and Parkinson's diseases including blood cells, skin fibroblast, proteomics, saliva, olfactory, stomach and colon, heart and peripheral nervous system, and others. Graphical Abstract.
Collapse
|
226
|
Yokoyama AS, Dunaway K, Rutkowsky J, Rutledge JC, Milenkovic D. Chronic consumption of a western diet modifies the DNA methylation profile in the frontal cortex of mice. Food Funct 2018; 9:1187-1198. [PMID: 29372223 DOI: 10.1039/c7fo01602f] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In our previous work in mice, we have shown that chronic consumption of a Western diet (WD; 42% kcal fat, 0.2% total cholesterol and 34% sucrose) is correlated with impaired cognitive function. Cognitive decline has also been associated with alterations in DNA methylation. Additionally, although there have been many studies analyzing the effect of maternal consumption of a WD on DNA methylation in the offspring, few studies have analyzed how an individual's consumption of a WD can impact his/her DNA methylation. Since the frontal cortex is involved in the regulation of cognitive function and is often affected in cases of cognitive decline, this study aimed to examine how chronic consumption of a WD affects DNA methylation in the frontal cortex of mice. Eight-week-old male mice were fed either a control diet (CD) or a WD for 12 weeks, after which time alterations in DNA methylation were analyzed. Assessment of global DNA methylation in the frontal cortex using dot blot analysis revealed that there was a decrease in global DNA methylation in the WD-fed mice compared with the CD-fed mice. Bioinformatic analysis identified several networks and pathways containing genes displaying differential methylation, particularly those involved in metabolism, cell adhesion and cytoskeleton integrity, inflammation and neurological function. In conclusion, the results from this study suggest that consumption of a WD alters DNA methylation in the frontal cortex of mice and could provide one of the mechanisms by which consumption of a WD impairs cognitive function.
Collapse
Affiliation(s)
- Amy S Yokoyama
- Department of Internal Medicine, Division of Cardiovascular Medicine, School of Medicine, University of California Davis, Davis, California 95616, USA.
| | | | | | | | | |
Collapse
|
227
|
Karch S, Broichhagen J, Schneider J, Böning D, Hartmann S, Schmid B, Tripal P, Palmisano R, Alzheimer C, Johnsson K, Huth T. A New Fluorogenic Small-Molecule Labeling Tool for Surface Diffusion Analysis and Advanced Fluorescence Imaging of β-Site Amyloid Precursor Protein-Cleaving Enzyme 1 Based on Silicone Rhodamine: SiR-BACE1. J Med Chem 2018; 61:6121-6139. [PMID: 29939737 DOI: 10.1021/acs.jmedchem.8b00387] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
β-site APP-cleaving enzyme 1 (BACE1) is a major player in the pathogenesis of Alzheimer's disease. Structural and functional fluorescence microscopy offers a powerful approach to learn about the physiology and pathophysiology of this protease. Up to now, however, common labeling techniques require genetic manipulation, use large antibodies, or are not compatible with live cell imaging. Fluorescent small molecules that specifically bind to the protein of interest can overcome these limitations. Herein, we introduce SiR-BACE1, a conjugate of the BACE1 inhibitor S-39 and SiR647, as a novel fluorogenic, tag-free, and antibody-free label for BACE1. We present its chemical development, characterize its photophysical and pharmacologic properties, and evaluate its behavior in solution, in overexpression systems, and in native brain tissue. We demonstrate its applicability in confocal, stimulated emission depletion and dynamic single-molecule microscopy. The first functional studies with SiR-BACE1 on the surface mobility of BACE1 revealed a markedly confined diffusion pattern.
Collapse
Affiliation(s)
- Sandra Karch
- Institute of Physiology and Pathophysiology , Friedrich-Alexander-Universität Erlangen-Nürnberg , Universitaetsstrasse 17 , 91054 Erlangen , Germany
| | - Johannes Broichhagen
- Department of Chemical Biology , Max Planck Institute for Medical Research , Jahnstrasse 29 , 69120 Heidelberg , Germany.,Laboratory of Protein Engineering, Institut des Sciences et Ingénierie Chimiques, Sciences de Base , École Polytechnique Fédérale Lausanne , 1015 Lausanne , Switzerland
| | - Julia Schneider
- Institute of Physiology and Pathophysiology , Friedrich-Alexander-Universität Erlangen-Nürnberg , Universitaetsstrasse 17 , 91054 Erlangen , Germany
| | - Daniel Böning
- Max Planck Institute for the Science of Light , Staudtstrasse 2 , 91058 Erlangen , Germany
| | - Stephanie Hartmann
- Institute of Physiology and Pathophysiology , Friedrich-Alexander-Universität Erlangen-Nürnberg , Universitaetsstrasse 17 , 91054 Erlangen , Germany
| | - Benjamin Schmid
- Optical Imaging Centre , Friedrich-Alexander-Universität Erlangen-Nürnberg , Hartmannstrasse 14 , 91052 Erlangen , Germany
| | - Philipp Tripal
- Optical Imaging Centre , Friedrich-Alexander-Universität Erlangen-Nürnberg , Hartmannstrasse 14 , 91052 Erlangen , Germany
| | - Ralf Palmisano
- Optical Imaging Centre , Friedrich-Alexander-Universität Erlangen-Nürnberg , Hartmannstrasse 14 , 91052 Erlangen , Germany
| | - Christian Alzheimer
- Institute of Physiology and Pathophysiology , Friedrich-Alexander-Universität Erlangen-Nürnberg , Universitaetsstrasse 17 , 91054 Erlangen , Germany
| | - Kai Johnsson
- Department of Chemical Biology , Max Planck Institute for Medical Research , Jahnstrasse 29 , 69120 Heidelberg , Germany.,Laboratory of Protein Engineering, Institut des Sciences et Ingénierie Chimiques, Sciences de Base , École Polytechnique Fédérale Lausanne , 1015 Lausanne , Switzerland
| | - Tobias Huth
- Institute of Physiology and Pathophysiology , Friedrich-Alexander-Universität Erlangen-Nürnberg , Universitaetsstrasse 17 , 91054 Erlangen , Germany
| |
Collapse
|
228
|
Huang M, Qi W, Fang S, Jiang P, Yang C, Mo Y, Dong C, Li Y, Zhong J, Cai W, Yang Z, Zhou T, Wang Q, Yang X, Gao G. Pigment Epithelium-Derived Factor Plays a Role in Alzheimer's Disease by Negatively Regulating Aβ42. Neurotherapeutics 2018; 15:728-741. [PMID: 29736859 PMCID: PMC6095778 DOI: 10.1007/s13311-018-0628-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. Pigment epithelium-derived factor (PEDF), a unique neurotrophic protein, decreases with aging. Previous reports have conflicted regarding whether the PEDF concentration is altered in AD patients. In addition, the effect of PEDF on AD has not been documented. Here, we tested serum samples of 31 AD patients and 271 normal controls. We found that compared to PEDF levels in young and middle-aged control subjects, PEDF levels were reduced in old-aged controls and even more so in AD patients. Furthermore, we verified that PEDF expression was much lower and amyloid β-protein (Aβ)42 expression was much higher in senescence-accelerated mouse prone 8 (SAMP8) strain mice than in senescence-accelerated mouse resistant 1 (SAMR1) control strain mice. Accordingly, high levels of Aβ42 were also observed in PEDF knockout (KO) mice. PEDF notably reduced cognitive impairment in the Morris water maze (MWM) and significantly downregulated Aβ42 in SAMP8 mice. Mechanistically, PEDF downregulated presenilin-1 (PS1) expression by inhibiting the c-Jun N-terminal kinase (JNK) pathway. Taken together, our findings demonstrate for the first time that PEDF negatively regulates Aβ42 and that PEDF deficiency with aging might play a crucial role in the development of AD.
Collapse
Affiliation(s)
- Mao Huang
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Weiwei Qi
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Shuhuan Fang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ping Jiang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Cong Yang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yousheng Mo
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chang Dong
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Yan Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Jun Zhong
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Weibin Cai
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Zhonghan Yang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Ti Zhou
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Xia Yang
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China.
- China Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China.
| | - Guoquan Gao
- Program of Molecular Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou, 510080, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
- Guangdong Engineering & Technology Research Center for Gene Manipulation and Biomacromolecular Products, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
229
|
Hayden KM, Inouye SK, Cunningham C, Jones RN, Avidan MS, Davis D, Kuchel GA, Tang Y, Khachaturian AS. Reduce the burden of dementia now. Alzheimers Dement 2018; 14:845-847. [PMID: 29959910 DOI: 10.1016/j.jalz.2018.06.3039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
| | - Sharon K Inouye
- Hebrew SeniorLife and Harvard Medical School, Boston, MA, USA
| | | | - Richard N Jones
- Alpert Medical School of Brown University, Providence, RI, USA
| | | | - Daniel Davis
- MRC Unit for Lifelong Health and Ageing at UCL, London, United Kingdom
| | | | - Yi Tang
- Xuan Wu Hospital, Capital Medical University, Beijing, China
| | | |
Collapse
|
230
|
Liao H, Zhu Z, Peng Y. Potential Utility of Retinal Imaging for Alzheimer's Disease: A Review. Front Aging Neurosci 2018; 10:188. [PMID: 29988470 PMCID: PMC6024140 DOI: 10.3389/fnagi.2018.00188] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 06/05/2018] [Indexed: 01/18/2023] Open
Abstract
The ensuing upward shift in demographic distribution due to the increase in life expectancy has resulted in a rising prevalence of Alzheimer's disease (AD). The heavy public burden of AD, along with the urgent to prevent and treat the disease before the irreversible damage to the brain, calls for a sensitive and specific screening technology to identify high-risk individuals before cognitive symptoms arise. Even though current modalities, such as positron emission tomography (PET) and cerebrospinal fluid (CSF) biomarker, showed their potential clinical uses in early detection of AD, the high cost, narrow isotope availability of PET probes and invasive characteristics of CSF biomarker limited their broad utility. Therefore, additional tools for detection of AD are needed. As a projection of the central nervous system (CNS), the retina has been described as a "window to the brain" and a novel marker for AD. Low cost, easy accessibility and non-invasive features make retina tests suitable for large-scale population screening and investigations of preclinical AD. Furthermore, a number of novel approaches in retina imaging, such as optical coherence tomography (OCT), have been developed and made it possible to visualize changes in the retina at a very fine resolution. In this review, we outline the background for AD to accelerate the adoption of retina imaging for the diagnosis and management of AD in clinical practice. Then, we focus on recent findings on the application of retina imaging to investigate AD and provide suggestions for future research directions.
Collapse
Affiliation(s)
- Huan Liao
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhuoting Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ying Peng
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
231
|
Cavanagh C, Wong TP. Preventing synaptic deficits in Alzheimer's disease by inhibiting tumor necrosis factor alpha signaling. IBRO Rep 2018; 4:18-21. [PMID: 30135948 PMCID: PMC6084902 DOI: 10.1016/j.ibror.2018.01.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/31/2018] [Indexed: 11/21/2022] Open
Abstract
The characterization of preclinical stages of Alzheimer's disease (AD) would provide a therapeutic window for prevention. One of the challenges of developing preventive therapy for AD is to identify early biomarkers for intervention studies. We have recently shown that in the TgCRND8 transgenic AD mouse model, increased hippocampal levels of the pro-inflammatory cytokine tumor necrosis factor alpha (TNFα) and enhanced excitatory synaptic transmission were early-onset changes that occurred weeks before amyloid plaque formation. Inhibiting TNFα before plaque formation not only normalized excitatory synaptic function, but also prevented the impairment of synaptic function 4 months later. In this review paper, we will examine the potential contributions of TNFα to the alteration of brain function in preclinical AD. The prospective use of TNFα inhibitors for preventing AD will be discussed.
Collapse
Affiliation(s)
- Chelsea Cavanagh
- Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Tak Pan Wong
- Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| |
Collapse
|
232
|
Lewczuk P, Riederer P, O’Bryant SE, Verbeek MM, Dubois B, Visser PJ, Jellinger KA, Engelborghs S, Ramirez A, Parnetti L, Jack CR, Teunissen CE, Hampel H, Lleó A, Jessen F, Glodzik L, de Leon MJ, Fagan AM, Molinuevo JL, Jansen WJ, Winblad B, Shaw LM, Andreasson U, Otto M, Mollenhauer B, Wiltfang J, Turner MR, Zerr I, Handels R, Thompson AG, Johansson G, Ermann N, Trojanowski JQ, Karaca I, Wagner H, Oeckl P, van Waalwijk van Doorn L, Bjerke M, Kapogiannis D, Kuiperij HB, Farotti L, Li Y, Gordon BA, Epelbaum S, Vos SJB, Klijn CJM, Van Nostrand WE, Minguillon C, Schmitz M, Gallo C, Mato AL, Thibaut F, Lista S, Alcolea D, Zetterberg H, Blennow K, Kornhuber J, Riederer P, Gallo C, Kapogiannis D, Mato AL, Thibaut F. Cerebrospinal fluid and blood biomarkers for neurodegenerative dementias: An update of the Consensus of the Task Force on Biological Markers in Psychiatry of the World Federation of Societies of Biological Psychiatry. World J Biol Psychiatry 2018; 19:244-328. [PMID: 29076399 PMCID: PMC5916324 DOI: 10.1080/15622975.2017.1375556] [Citation(s) in RCA: 196] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the 12 years since the publication of the first Consensus Paper of the WFSBP on biomarkers of neurodegenerative dementias, enormous advancement has taken place in the field, and the Task Force takes now the opportunity to extend and update the original paper. New concepts of Alzheimer's disease (AD) and the conceptual interactions between AD and dementia due to AD were developed, resulting in two sets for diagnostic/research criteria. Procedures for pre-analytical sample handling, biobanking, analyses and post-analytical interpretation of the results were intensively studied and optimised. A global quality control project was introduced to evaluate and monitor the inter-centre variability in measurements with the goal of harmonisation of results. Contexts of use and how to approach candidate biomarkers in biological specimens other than cerebrospinal fluid (CSF), e.g. blood, were precisely defined. Important development was achieved in neuroimaging techniques, including studies comparing amyloid-β positron emission tomography results to fluid-based modalities. Similarly, development in research laboratory technologies, such as ultra-sensitive methods, raises our hopes to further improve analytical and diagnostic accuracy of classic and novel candidate biomarkers. Synergistically, advancement in clinical trials of anti-dementia therapies energises and motivates the efforts to find and optimise the most reliable early diagnostic modalities. Finally, the first studies were published addressing the potential of cost-effectiveness of the biomarkers-based diagnosis of neurodegenerative disorders.
Collapse
Affiliation(s)
- Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, and Department of Biochemical Diagnostics, University Hospital of Białystok, Białystok, Poland
| | - Peter Riederer
- Center of Mental Health, Clinic and Policlinic of Psychiatry, Psychosomatics and Psychotherapy, University Hospital Würzburg, Würzburg, Germany
| | - Sid E. O’Bryant
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Marcel M. Verbeek
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Center, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer center, Nijmegen, The Netherlands
| | - Bruno Dubois
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Salpêtrièrie Hospital, INSERM UMR-S 975 (ICM), Paris 6 University, Paris, France
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
- Department of Neurology, Alzheimer Centre, Amsterdam Neuroscience VU University Medical Centre, Amsterdam, The Netherlands
| | | | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), University of Antwerp, Antwerp, Belgium
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Alfredo Ramirez
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | - Lucilla Parnetti
- Section of Neurology, Center for Memory Disturbances, Lab of Clinical Neurochemistry, University of Perugia, Perugia, Italy
| | | | - Charlotte E. Teunissen
- Neurochemistry Lab and Biobank, Department of Clinical Chemistry, Amsterdam Neuroscience, VU University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - Harald Hampel
- AXA Research Fund & UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
| | - Alberto Lleó
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Spain
| | - Frank Jessen
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
- German Center for Neurodegenerative Disorders (DZNE), Bonn, Germany
| | - Lidia Glodzik
- Center for Brain Health, Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA
| | - Mony J. de Leon
- Center for Brain Health, Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA
| | - Anne M. Fagan
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | - José Luis Molinuevo
- Barcelonabeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
- Alzheimer’s Disease and Other Cognitive Disorders Unit, Hospital Clínic, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Willemijn J. Jansen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
| | - Bengt Winblad
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Huddinge, Sweden
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ulf Andreasson
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik, Kassel and University Medical Center Göttingen, Department of Neurology, Göttingen, Germany
| | - Jens Wiltfang
- Department of Psychiatry & Psychotherapy, University of Göttingen, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- iBiMED, Medical Sciences Department, University of Aveiro, Aveiro, Portugal
| | - Martin R. Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Inga Zerr
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Clinical Dementia Centre, Department of Neurology, University Medical School, Göttingen, Germany
| | - Ron Handels
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Huddinge, Sweden
| | | | - Gunilla Johansson
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Huddinge, Sweden
| | - Natalia Ermann
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - John Q. Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilker Karaca
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Holger Wagner
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Patrick Oeckl
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Linda van Waalwijk van Doorn
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Center, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer center, Nijmegen, The Netherlands
| | - Maria Bjerke
- Reference Center for Biological Markers of Dementia (BIODEM), University of Antwerp, Antwerp, Belgium
| | - Dimitrios Kapogiannis
- Laboratory of Neurosciences, National Institute on Aging/National Institutes of Health (NIA/NIH), Baltimore, MD, USA
| | - H. Bea Kuiperij
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Center, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer center, Nijmegen, The Netherlands
| | - Lucia Farotti
- Section of Neurology, Center for Memory Disturbances, Lab of Clinical Neurochemistry, University of Perugia, Perugia, Italy
| | - Yi Li
- Center for Brain Health, Department of Psychiatry, NYU Langone Medical Center, New York, NY, USA
| | - Brian A. Gordon
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Stéphane Epelbaum
- Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Salpêtrièrie Hospital, INSERM UMR-S 975 (ICM), Paris 6 University, Paris, France
| | - Stephanie J. B. Vos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
| | - Catharina J. M. Klijn
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Radboud Alzheimer Center, Nijmegen, The Netherlands
| | | | - Carolina Minguillon
- Barcelonabeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | - Matthias Schmitz
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Clinical Dementia Centre, Department of Neurology, University Medical School, Göttingen, Germany
| | - Carla Gallo
- Departamento de Ciencias Celulares y Moleculares/Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Andrea Lopez Mato
- Chair of Psychoneuroimmunoendocrinology, Maimonides University, Buenos Aires, Argentina
| | - Florence Thibaut
- Department of Psychiatry, University Hospital Cochin-Site Tarnier 89 rue d’Assas, INSERM 894, Faculty of Medicine Paris Descartes, Paris, France
| | - Simone Lista
- AXA Research Fund & UPMC Chair, Sorbonne Universités, Université Pierre et Marie Curie (UPMC) Paris 06, Inserm, CNRS, Institut du Cerveau et de la Moelle Épinière (ICM), Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Boulevard de l’hôpital, Paris, France
| | - Daniel Alcolea
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, CIBERNED, Spain
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | | | | | |
Collapse
|
233
|
Santos CDSD, Bessa TAD, Xavier AJ. Factors associated with dementia in elderly. CIENCIA & SAUDE COLETIVA 2018; 25:603-611. [PMID: 32022200 DOI: 10.1590/1413-81232020252.02042018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 05/16/2018] [Indexed: 12/29/2022] Open
Abstract
We analyzed the factors associated with dementia in the elderly attended at a memory outpatient clinic of the University of Southern Santa Catarina (UNISUL). This is a cross-sectional study with data analysis of medical records from January 2013 to April 2016. The outcome was the clinical diagnosis of dementia. The control variables were: serum vitamin D level at the time of diagnosis, gender, skin color, schooling, age, type 2 diabetes, hypertension, and depression. We performed a crude and adjusted analysis with logistic regression. The sample consisted of 287 elderly, with the predominance of age between 60 and 69 years (48.78%), female (79.09%) and white (92.33%). The mean number of years of study was 6.95 years (SD ± 4.95) and mean vitamin D was 26.09 ng/mL (SD ± 9,20). The prevalence of elderly with dementia was 16.72%. Depression was the most prevalent (42.50%) among the morbidities, followed by hypertension (31.71%). The following were independently associated with dementia: vitamin D (OR = 0.92, 95%CI, 0.88;0.97), depression (OR = 4.09, 95%CI, 1.87;8.94), hypertension (OR = 2.65, 95%CI, 1.15;6.08) and individuals aged 80 years and over (OR = 3.97 95%CI, 1.59;9.91). Dementia prevalence was high and diagnosed dementia was associated with lower levels of vitamin D. Vitamin D is a modifiable factor, opening up essential perspectives for public health policies.
Collapse
Affiliation(s)
- Camila de Souza Dos Santos
- Universidade Federal de Santa Catarina. R. Eng. Agrônomo Andrei Cristian Ferreira s/n, Trindade. 88040-900, Florianópolis, SC, Brasil.
| | - Thaíssa Araujo de Bessa
- Universidade Federal de Santa Catarina. R. Eng. Agrônomo Andrei Cristian Ferreira s/n, Trindade. 88040-900, Florianópolis, SC, Brasil.
| | - André Junqueira Xavier
- Universidade Federal de Santa Catarina. R. Eng. Agrônomo Andrei Cristian Ferreira s/n, Trindade. 88040-900, Florianópolis, SC, Brasil.
| |
Collapse
|
234
|
Anderson-Hanley C, Barcelos NM, Zimmerman EA, Gillen RW, Dunnam M, Cohen BD, Yerokhin V, Miller KE, Hayes DJ, Arciero PJ, Maloney M, Kramer AF. The Aerobic and Cognitive Exercise Study (ACES) for Community-Dwelling Older Adults With or At-Risk for Mild Cognitive Impairment (MCI): Neuropsychological, Neurobiological and Neuroimaging Outcomes of a Randomized Clinical Trial. Front Aging Neurosci 2018; 10:76. [PMID: 29780318 PMCID: PMC5945889 DOI: 10.3389/fnagi.2018.00076] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 03/07/2018] [Indexed: 12/19/2022] Open
Abstract
Prior research has found that cognitive benefits of physical exercise and brain health in older adults may be enhanced when mental exercise is interactive simultaneously, as in exergaming. It is unclear whether the cognitive benefit can be maximized by increasing the degree of mental challenge during exercise. This randomized clinical trial (RCT), the Aerobic and Cognitive Exercise Study (ACES) sought to replicate and extend prior findings of added cognitive benefit from exergaming to those with or at risk for mild cognitive impairment (MCI). ACES compares the effects of 6 months of an exer-tour (virtual reality bike rides) with the effects of a more effortful exer-score (pedaling through a videogame to score points). Fourteen community-dwelling older adults meeting screening criteria for MCI (sMCI) were adherent to their assigned exercise for 6 months. The primary outcome was executive function, while secondary outcomes included memory and everyday cognitive function. Exer-tour and exer-score yielded significant moderate effects on executive function (Stroop A/C; d's = 0.51 and 0.47); there was no significant interaction effect. However, after 3 months the exer-tour revealed a significant and moderate effect, while exer-score showed little impact, as did a game-only condition. Both exer-tour and exer-score conditions also resulted in significant improvements in verbal memory. Effects appear to generalize to self-reported everyday cognitive function. Pilot data, including salivary biomarkers and structural MRI, were gathered at baseline and 6 months; exercise dose was associated with increased BDNF as well as increased gray matter volume in the PFC and ACC. Improvement in memory was associated with an increase in the DLPFC. Improved executive function was associated with increased expression of exosomal miRNA-9. Interactive physical and cognitive exercise (both high and low mental challenge) yielded similarly significant cognitive benefit for adherent sMCI exercisers over 6 months. A larger RCT is needed to confirm these findings. Further innovation and clinical trial data are needed to develop accessible, yet engaging and effective interventions to combat cognitive decline for the growing MCI population. ClinicalTrials.gov ID: NCT02237560
Collapse
Affiliation(s)
- Cay Anderson-Hanley
- The Healthy Aging and Neuropsychology Lab, Union College, Schenectady, NY, United States
| | - Nicole M Barcelos
- The Healthy Aging and Neuropsychology Lab, Union College, Schenectady, NY, United States
| | - Earl A Zimmerman
- Alzheimer's Disease Center, Albany Medical Center, Albany, NY, United States
| | - Robert W Gillen
- Sunnyview Rehabilitation Hospital, Schenectady, NY, United States
| | - Mina Dunnam
- Stratton VA Medical Center, Albany, NY, United States
| | - Brian D Cohen
- Department of Biology, Union College, Schenectady, NY, United States
| | - Vadim Yerokhin
- Biomedical Sciences Department, Oklahoma State University, Tulsa, OK, United States
| | - Kenneth E Miller
- Department of Anatomy and Cell Biology, Oklahoma State University, Tulsa, OK, United States
| | - David J Hayes
- The Healthy Aging and Neuropsychology Lab, Union College, Schenectady, NY, United States
| | - Paul J Arciero
- Department of Health & Human Physiological Sciences, Skidmore College, Saratoga Springs, NY, United States
| | - Molly Maloney
- The Healthy Aging and Neuropsychology Lab, Union College, Schenectady, NY, United States
| | - Arthur F Kramer
- Beckman Institute, University of Illinois, Urbana-Champaign, Champaign, IL, United States
| |
Collapse
|
235
|
Fisher DW, Bennett DA, Dong H. Sexual dimorphism in predisposition to Alzheimer's disease. Neurobiol Aging 2018; 70:308-324. [PMID: 29754747 DOI: 10.1016/j.neurobiolaging.2018.04.004] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 04/08/2018] [Accepted: 04/10/2018] [Indexed: 12/20/2022]
Abstract
Clinical studies indicate that Alzheimer's disease (AD) disproportionately affects women in both disease prevalence and rate of symptom progression, but the mechanisms underlying this sexual divergence are unknown. Although some have suggested this difference in risk is a reflection of the known differences in longevity between men and women, mounting clinical and preclinical evidence supports women also having intrinsic susceptibilities toward the disease. Although a number of potential risk factors have been hypothesized to mediate these differences, none have been definitively verified. In this review, we first summarize the epidemiologic studies of prevalence and incidence of AD among the sexes. Next, we discuss the most likely risk factors to date that interact with biological sex, including (1) genetic factors, (2) sex hormones (3) deviations in brain structure, (4) inflammation and microglia, and (5) and psychosocial stress responses. Overall, though differences in life span are likely to account for part of the divide between the sexes in AD prevalence, the abundance of preclinical and clinical evidence presented here suggests an increase in intrinsic AD risk for women. Therefore, future studies focusing on the underlying biological mechanisms for this phenomenon are needed to better understand AD pathogenesis in both sexes, with the eventual goal of sex-specific prevention and treatment strategies.
Collapse
Affiliation(s)
- Daniel W Fisher
- Departments of Psychiatry and Behavioral Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - David A Bennett
- Department of Neurological Sciences, Rush Alzheimer's Disease Center, Rush Medical College, Chicago, IL, USA
| | - Hongxin Dong
- Departments of Psychiatry and Behavioral Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
236
|
Erkkinen MG, Kim MO, Geschwind MD. Clinical Neurology and Epidemiology of the Major Neurodegenerative Diseases. Cold Spring Harb Perspect Biol 2018; 10:a033118. [PMID: 28716886 PMCID: PMC5880171 DOI: 10.1101/cshperspect.a033118] [Citation(s) in RCA: 665] [Impact Index Per Article: 95.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Neurodegenerative diseases are a common cause of morbidity and cognitive impairment in older adults. Most clinicians who care for the elderly are not trained to diagnose these conditions, perhaps other than typical Alzheimer's disease (AD). Each of these disorders has varied epidemiology, clinical symptomatology, laboratory and neuroimaging features, neuropathology, and management. Thus, it is important that clinicians be able to differentiate and diagnose these conditions accurately. This review summarizes and highlights clinical aspects of several of the most commonly encountered neurodegenerative diseases, including AD, frontotemporal dementia (FTD) and its variants, progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), Parkinson's disease (PD), dementia with Lewy bodies (DLB), multiple system atrophy (MSA), and Huntington's disease (HD). For each condition, we provide a brief overview of the epidemiology, defining clinical symptoms and diagnostic criteria, relevant imaging and laboratory features, genetics, pathology, treatments, and differential diagnosis.
Collapse
Affiliation(s)
- Michael G Erkkinen
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, California 94158
| | - Mee-Ohk Kim
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, California 94158
| | - Michael D Geschwind
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, California 94158
| |
Collapse
|
237
|
Hunter JC, Handing EP, Casanova R, Kuchibhatla M, Lutz MW, Saldana S, Plassman BL, Hayden KM. Neighborhoods, sleep quality, and cognitive decline: Does where you live and how well you sleep matter? Alzheimers Dement 2018; 14:454-461. [PMID: 29396109 PMCID: PMC5899662 DOI: 10.1016/j.jalz.2017.10.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 09/27/2017] [Accepted: 10/12/2017] [Indexed: 11/21/2022]
Abstract
INTRODUCTION We evaluated the association between neighborhood socioeconomic status (NSES) and sleep quality on cognitive decline in the Health and Retirement Study. METHODS Health and Retirement Study participants (n = 8090), aged 65+ with DNA and multiple biennial cognitive observations (abbreviated Telephone Interview for Cognitive Status), were included. Participants were grouped into quartiles of NSES and sleep quality scores. We adjusted for apolipoprotein E ε4, demographic, and cardiovascular risk factors. Random effects modeling evaluated cognitive change over time. RESULTS NSES and sleep were significantly associated with cognitive decline, and there was a significant interaction between them (P = .02). Significant differences between high/low NSES and high/low sleep quality (P < .0001) were found. DISCUSSION Sleep and NSES were associated with cognitive decline; the association between sleep and cognition appeared stronger among those with low NSES. The association between low NSES, poor sleep quality, and cognitive decline was roughly equivalent to the association between apolipoprotein E ε4 and cognitive decline.
Collapse
Affiliation(s)
- Jaimie C Hunter
- Department of Social Sciences and Health Policy, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Elizabeth P Handing
- Department of Internal Medicine, Section on Geriatrics and Gerontology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Ramon Casanova
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Maragatha Kuchibhatla
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, USA
| | - Michael W Lutz
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Santiago Saldana
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Brenda L Plassman
- Department of Neurology, Duke University Medical Center, Durham, NC, USA; Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Kathleen M Hayden
- Department of Social Sciences and Health Policy, Wake Forest School of Medicine, Winston-Salem, NC, USA; Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
238
|
Doniger GM, Beeri MS, Bahar-Fuchs A, Gottlieb A, Tkachov A, Kenan H, Livny A, Bahat Y, Sharon H, Ben-Gal O, Cohen M, Zeilig G, Plotnik M. Virtual reality-based cognitive-motor training for middle-aged adults at high Alzheimer's disease risk: A randomized controlled trial. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2018; 4:118-129. [PMID: 29955655 PMCID: PMC6021455 DOI: 10.1016/j.trci.2018.02.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Introduction Ubiquity of Alzheimer's disease (AD) coupled with relatively ineffectual pharmacologic treatments has spurred interest in nonpharmacologic lifestyle interventions for prevention or risk reduction. However, evidence of neuroplasticity notwithstanding, there are few scientifically rigorous, ecologically relevant brain training studies focused on building cognitive reserve in middle age to protect against cognitive decline. This pilot study will examine the ability of virtual reality (VR) cognitive training to improve cognition and cerebral blood flow (CBF) in middle-aged individuals at high AD risk due to parental history. Methods The design is an assessor-blind, parallel group, randomized controlled trial of VR cognitive-motor training in middle-aged adults with AD family history. The experimental group will be trained with adaptive “real-world” VR tasks targeting sustained and selective attention, working memory, covert rule deduction, and planning, while walking on a treadmill. One active control group will perform the VR tasks without treadmill walking; another will walk on a treadmill while watching scientific documentaries (nonspecific cognitive stimulation). A passive (waitlist) control group will not receive training. Training sessions will be 45 minutes, twice/week for 12 weeks. Primary outcomes are global cognition and CBF (from arterial spin labeling [ASL]) at baseline, immediately after training (training gain), and 3 months post-training (maintenance gain). We aim to recruit 125 participants, including 20 passive controls and 35 in the other groups. Discussion Current pharmacologic therapies are for symptomatic AD patients, whereas nonpharmacologic training is administrable before symptom onset. Emerging evidence suggests that cognitive training improves cognitive function. However, a more ecologically valid cognitive-motor VR setting that better mimics complex daily activities may augment transfer of trained skills. VR training has benefited clinical cohorts, but benefit in asymptomatic high-risk individuals is unknown. If effective, this trial may help define a prophylactic regimen for AD, adaptable for home-based application in high-risk individuals.
Collapse
Affiliation(s)
- Glen M. Doniger
- Center of Advanced Technologies in Rehabilitation, Sheba Medical Center, Ramat Gan, Israel
- Joseph Sagol Neuroscience Center, Sheba Medical Center, Ramat Gan, Israel
- Baruch Ivcher School of Psychology, Interdisciplinary Center (IDC) Herzliya, Herzliya, Israel
- Corresponding author. Tel.: +97235304874; Fax: +97235307572.
| | - Michal Schnaider Beeri
- Joseph Sagol Neuroscience Center, Sheba Medical Center, Ramat Gan, Israel
- Baruch Ivcher School of Psychology, Interdisciplinary Center (IDC) Herzliya, Herzliya, Israel
- Department of Psychiatry, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alex Bahar-Fuchs
- Joseph Sagol Neuroscience Center, Sheba Medical Center, Ramat Gan, Israel
- Center for Research on Aging, Health, and Wellbeing, Research School of Population Health, The Australian National University, Canberra, ACT, Australia
- The Academic Unit for Psychiatry of Old Age, Department of Psychiatry, The University of Melbourne, Victoria, Australia
| | - Amihai Gottlieb
- Center of Advanced Technologies in Rehabilitation, Sheba Medical Center, Ramat Gan, Israel
| | - Anastasia Tkachov
- Joseph Sagol Neuroscience Center, Sheba Medical Center, Ramat Gan, Israel
| | - Hagar Kenan
- Joseph Sagol Neuroscience Center, Sheba Medical Center, Ramat Gan, Israel
| | - Abigail Livny
- Joseph Sagol Neuroscience Center, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Diagnostic Imaging, Sheba Medical Center, Ramat Gan, Israel
| | - Yotam Bahat
- Center of Advanced Technologies in Rehabilitation, Sheba Medical Center, Ramat Gan, Israel
| | - Hadar Sharon
- Center of Advanced Technologies in Rehabilitation, Sheba Medical Center, Ramat Gan, Israel
| | - Oran Ben-Gal
- Center of Advanced Technologies in Rehabilitation, Sheba Medical Center, Ramat Gan, Israel
| | - Maya Cohen
- Center of Advanced Technologies in Rehabilitation, Sheba Medical Center, Ramat Gan, Israel
- Joseph Sagol Neuroscience Center, Sheba Medical Center, Ramat Gan, Israel
| | - Gabi Zeilig
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Neurological Rehabilitation, Sheba Medical Center, Ramat Gan, Israel
| | - Meir Plotnik
- Center of Advanced Technologies in Rehabilitation, Sheba Medical Center, Ramat Gan, Israel
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
239
|
Kim J, Kim H, Roh H, Kwon Y. Causes of hyperhomocysteinemia and its pathological significance. Arch Pharm Res 2018; 41:372-383. [PMID: 29552692 DOI: 10.1007/s12272-018-1016-4] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 02/26/2018] [Indexed: 02/03/2023]
Abstract
In the last 10 years, homocysteine has been regarded as a marker of cardiovascular disease and a definite risk factor for many other diseases. Homocysteine is biosynthesized from methionine through multiple steps and then goes through one of two major metabolic pathways: remethylation and transsulfuration. Hyperhomocysteinemia is a state in which too much homocysteine is present in the body. The main cause of hyperhomocysteinemia is a dysfunction of enzymes and cofactors associated with the process of homocysteine biosynthesis. Other causes include excessive methionine intake, certain diseases and side effects of some drugs. Hyperhomocysteinemia is a trigger for many diseases, such as atherosclerosis, congestive heart failure, age-related macular degeneration, Alzheimer's disease and hearing loss. There are many studies showing a positive relationship between homocysteine level and various symptoms. We speculate that a high level of homocysteine can be the sole reason or an aggravating factor in numerous diseases for which causal links are not fully understood.
Collapse
Affiliation(s)
- Jihyun Kim
- Ewha Institute for Global Pharmacy Leadership, College of Pharmacy, Ewha Womans University, Seoul, 120-750, Republic of Korea
| | - Hyunhee Kim
- Ewha Institute for Global Pharmacy Leadership, College of Pharmacy, Ewha Womans University, Seoul, 120-750, Republic of Korea
| | - Heewon Roh
- Ewha Institute for Global Pharmacy Leadership, College of Pharmacy, Ewha Womans University, Seoul, 120-750, Republic of Korea
| | - Youngjoo Kwon
- Ewha Institute for Global Pharmacy Leadership, College of Pharmacy, Ewha Womans University, Seoul, 120-750, Republic of Korea.
| |
Collapse
|
240
|
Tariot PN, Lopera F, Langbaum JB, Thomas RG, Hendrix S, Schneider LS, Rios-Romenets S, Giraldo M, Acosta N, Tobon C, Ramos C, Espinosa A, Cho W, Ward M, Clayton D, Friesenhahn M, Mackey H, Honigberg L, Sanabria Bohorquez S, Chen K, Walsh T, Langlois C, Reiman EM. The Alzheimer's Prevention Initiative Autosomal-Dominant Alzheimer's Disease Trial: A study of crenezumab versus placebo in preclinical PSEN1 E280A mutation carriers to evaluate efficacy and safety in the treatment of autosomal-dominant Alzheimer's disease, including a placebo-treated noncarrier cohort. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2018; 4:150-160. [PMID: 29955659 PMCID: PMC6021543 DOI: 10.1016/j.trci.2018.02.002] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
INTRODUCTION Autosomal-dominant Alzheimer's disease (ADAD) represents a crucial population for identifying prevention strategies that might modify disease course for cognitively unimpaired individuals at high imminent risk for developing symptoms due to Alzheimer's disease (AD), that is, who have "preclinical" AD. Crenezumab is an antiamyloid monoclonal antibody that binds monomeric and aggregated forms of amyloid β, with highest affinity for oligomers; it is in development for early stages of sporadic AD and for ADAD. METHODS This is a prospective, randomized, double-blind, placebo-controlled phase 2 study of the efficacy of crenezumab versus placebo in asymptomatic PSEN1 E280A mutation carriers from family kindreds with ADAD in Colombia. Participants were randomized to receive either crenezumab or placebo for 260 weeks. The study was designed to enroll a planned total of 300 participants, including 200 preclinical mutation carriers (approximately 100 treatment, 100 placebo) and an additional control group of mutation noncarriers from the same family kindreds included to mask mutation carrier status (100 placebo only). The primary outcome is change in the Alzheimer's Prevention Initiative ADAD Composite Cognitive Test Score from baseline to week 260. Secondary outcomes include time to progression to mild cognitive impairment due to AD or dementia due to AD; changes in dementia severity, memory, and overall neurocognitive functioning; and changes in amyloid-positron emission tomography, fluorodeoxyglucose-positron emission tomography, magnetic resonance imaging volumes, and cerebrospinal fluid levels of β amyloid, tau, and p-tau. Safety and tolerability are assessed. RESULTS Two hundred fifty-two participants were enrolled between December 2013 and February 2017. DISCUSSION We describe the first large-scale, potentially label-enabling clinical trial of a preclinical treatment for ADAD. Results from this trial will inform on the efficacy of crenezumab for delaying onset of, slowing decline in, or preventing cognitive impairment in individuals with preclinical ADAD and will foster an improved understanding of AD biomarkers and their relationship to clinical outcomes.
Collapse
Affiliation(s)
| | - Francisco Lopera
- Grupo de Neurociencias, Universidad de Antioquia, SIU, Medellín, Colombia
| | | | - Ronald G. Thomas
- Department of Neurosciences, University of California San Diego School of Medicine, La Jolla, CA, USA
| | | | - Lon S. Schneider
- USC State of California Alzheimer's Disease Research and Clinical Center, Keck Medicine of USC, Los Angeles, CA, USA
| | | | - Margarita Giraldo
- Grupo de Neurociencias, Universidad de Antioquia, SIU, Medellín, Colombia
| | - Natalia Acosta
- Grupo de Neurociencias, Universidad de Antioquia, SIU, Medellín, Colombia
| | - Carlos Tobon
- Grupo de Neurociencias, Universidad de Antioquia, SIU, Medellín, Colombia
| | - Claudia Ramos
- Grupo de Neurociencias, Universidad de Antioquia, SIU, Medellín, Colombia
| | - Alejandro Espinosa
- Grupo de Neurociencias, Universidad de Antioquia, SIU, Medellín, Colombia
| | - William Cho
- Genentech, a Member of the Roche Group, South San Francisco, CA, USA
| | - Michael Ward
- Genentech, a Member of the Roche Group, South San Francisco, CA, USA
| | - David Clayton
- Genentech, a Member of the Roche Group, South San Francisco, CA, USA
| | | | - Howard Mackey
- Genentech, a Member of the Roche Group, South San Francisco, CA, USA
| | - Lee Honigberg
- Genentech, a Member of the Roche Group, South San Francisco, CA, USA
| | | | - Kewei Chen
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| | | | | | | | | |
Collapse
|
241
|
Aldehri M, Temel Y, Alnaami I, Jahanshahi A, Hescham S. Deep brain stimulation for Alzheimer's Disease: An update. Surg Neurol Int 2018; 9:58. [PMID: 29576909 PMCID: PMC5858049 DOI: 10.4103/sni.sni_342_17] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 01/09/2018] [Indexed: 12/23/2022] Open
Abstract
Background: Dementia is among the leading causes of severe and long-term disability worldwide, decreasing the quality of life of individuals and families. Moreover, it induces an enormous economic burden on societies. The most prevalent cause of dementia is Alzheimer's disease (AD). Because current treatment options for AD are limited, deep brain stimulation (DBS) has been considered. Methods: The aim of this review is to survey the current understanding regarding the effects of DBS in AD and possibly shed light on the mechanisms of DBS in AD. We searched PubMed and Cochrane for various studies in English literature describing DBS in patients with AD and relevant preclinical studies. All related studies published from December 2013 to March 2017 were included in this review. Results: Our understanding of the neural circuitry underlying learning and memory in both rodent models and human patients has grown over the past years and provided potential therapeutic targets for DBS such as the fornix and the nucleus basalis of Meynert. Clinical results indicate that DBS is most beneficial for patients who are in the early stages of AD. Potential mechanisms of action of DBS in AD comprise long-term structural plasticity, including hippocampal enlargement as well as enhanced neurotransmitter release. Conclusion: It is still premature to conclude that DBS can be used in the treatment of AD, and the field will wait for the results of ongoing and future clinical trials.
Collapse
Affiliation(s)
- Majed Aldehri
- Department of Neurosurgery, Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands.,Division of Neuroanatomy, Department of Anatomy, King Khalid University, Abha, Saudi Arabia.,European Graduate School of Neuroscience (EURON), Maastricht University, Maastricht, The Netherlands
| | - Yasin Temel
- Department of Neurosurgery, Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Ibrahim Alnaami
- Division of Neurosurgery, Department of Surgery, King Khalid University, Abha, Saudi Arabia
| | - Ali Jahanshahi
- Department of Neurosurgery, Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Sarah Hescham
- Department of Neurosurgery, Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
242
|
Soldan A, Pettigrew C, Albert M. Evaluating Cognitive Reserve Through the Prism of Preclinical Alzheimer Disease. Psychiatr Clin North Am 2018; 41:65-77. [PMID: 29412849 PMCID: PMC5806143 DOI: 10.1016/j.psc.2017.10.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The concept of cognitive reserve (CR) was proposed to account for the discrepancy between levels of brain pathologic features or damage and clinical and cognitive function. This article provides a detailed review of prospective longitudinal studies that have investigated the interaction between CR and Alzheimer's disease (AD) biomarkers on clinical and cognitive outcomes among individuals with preclinical AD. Current evidence shows that higher levels of CR are associated with a delay in the onset of symptoms of mild cognitive impairment and that there may be multiple pathways by which CR exerts its protective effects.
Collapse
Affiliation(s)
- Anja Soldan
- Department of Neurology, Johns Hopkins School of Medicine, 1620 McElderry Street, Reed Hall West - 1, Baltimore, MD 21205, USA.
| | - Corinne Pettigrew
- Research Associate, Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Marilyn Albert
- Professor, Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
243
|
|
244
|
Brookmeyer R, Abdalla N, Kawas CH, Corrada MM. Forecasting the prevalence of preclinical and clinical Alzheimer's disease in the United States. Alzheimers Dement 2018; 14:121-129. [PMID: 29233480 PMCID: PMC5803316 DOI: 10.1016/j.jalz.2017.10.009] [Citation(s) in RCA: 281] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 09/27/2017] [Accepted: 10/12/2017] [Indexed: 01/02/2023]
Abstract
INTRODUCTION We forecast the prevalence of preclinical and clinical Alzheimer's disease (AD) and evaluated potential impacts of primary and secondary preventions in the United States. METHODS We used a multistate model incorporating biomarkers for preclinical AD with US population projections. RESULTS Approximately 6.08 million Americans had either clinical AD or mild cognitive impairment due to AD in 2017 and that will grow to 15.0 million by 2060. In 2017, 46.7 million Americans had preclinical AD (amyloidosis, neurodegeneration, or both), although many may not progress to clinical disease during their lifetimes. Primary and secondary preventions have differential impact on future disease burden. DISCUSSION Because large numbers of persons are living with preclinical AD, our results underscore the need for secondary preventions for persons with existing AD brain pathology who are likely to develop clinical disease during their lifetimes as well as primary preventions for persons without preclinical disease.
Collapse
Affiliation(s)
- Ron Brookmeyer
- Department of Biostatistics, University of California, Los Angeles, CA 90095,
| | - Nada Abdalla
- Department of Biostatistics, University of California, Los Angeles, CA 90095,
| | - Claudia H. Kawas
- Departments of Neurology, Neurobiology and Behavior, Epidemiology and Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697,
| | - María M. Corrada
- Departments of Neurology, Epidemiology and Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697,
| |
Collapse
|
245
|
Yan Y, Dominguez S, Fisher DW, Dong H. Sex differences in chronic stress responses and Alzheimer's disease. Neurobiol Stress 2018; 8:120-126. [PMID: 29888307 PMCID: PMC5991323 DOI: 10.1016/j.ynstr.2018.03.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/10/2018] [Accepted: 03/17/2018] [Indexed: 11/13/2022] Open
Abstract
Clinical studies indicate that Alzheimer's disease (AD) disproportionately affects women in both disease prevalence and severity, but the mechanisms underlying this sex divergence are unknown. Though some have suggested this difference in risk is a reflection of known differences in longevity between men and women, mounting clinical and preclinical evidence supports women also having intrinsic susceptibilities towards the disease. While a number of potential risk factors have been hypothesized to affect these differences in risks, none have been definitively verified. In this review, we discuss a novel hypothesis whereby women's susceptibility to chronic stress also mediates increased risk for AD. As stress is a risk factor for AD, and women are twice as likely to develop mood disorders where stress is a major etiology, it is possible that sex dimorphisms in stress responses contribute to the increase in women with AD. In line with this, sex divergence in biochemical responses to stress have been noted along the hypothalamic-pituitary-adrenal (HPA) axis and among known molecular effectors of AD, with crosstalk between these processes also being likely. In addition, activation of the cortical corticotrophin-releasing factor receptor 1 (CRF1) signaling pathway leads to distinct female-biased increases in molecules associated with AD pathogenesis. Therefore, the different biochemical responses to stress between women and men may represent an intrinsic, sex-dependent risk factor for AD.
Collapse
Affiliation(s)
- Yan Yan
- Department of Psychiatry & Behavioral Sciences, Northwestern University, Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
- Department of Physiology, Zunyi Medical University, Zunyi Guizhou 563099, China
| | - Sky Dominguez
- Department of Psychiatry & Behavioral Sciences, Northwestern University, Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Daniel W. Fisher
- Department of Neurology, Northwestern University, Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Hongxin Dong
- Department of Psychiatry & Behavioral Sciences, Northwestern University, Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
- Department of Physiology, Zunyi Medical University, Zunyi Guizhou 563099, China
| |
Collapse
|
246
|
Oxidant/Antioxidant Imbalance in Alzheimer's Disease: Therapeutic and Diagnostic Prospects. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6435861. [PMID: 29636850 PMCID: PMC5831771 DOI: 10.1155/2018/6435861] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 12/18/2017] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and a great socioeconomic burden in the aging society. Compelling evidence demonstrates that molecular change characteristics for AD, such as oxidative stress and amyloid β (Aβ) oligomerization, precede by decades the onset of clinical dementia and that the disease represents a biological and clinical continuum of stages, from asymptomatic to severely impaired. Nevertheless, the sequence of the early molecular alterations and the interplay between them are incompletely understood. This review presents current knowledge about the oxidative stress-induced impairments and compromised oxidative stress defense mechanisms in AD brain and the cross-talk between various pathophysiological insults, with the focus on excessive reactive oxygen species (ROS) generation and Aβ overproduction at the early stages of the disease. Prospects for AD therapies targeting oxidant/antioxidant imbalance are being discussed, as well as for the development of novel oxidative stress-related, blood-based biomarkers for early, noninvasive AD diagnostics.
Collapse
|
247
|
Wang JH, Cheng XR, Zhang XR, Wang TX, Xu WJ, Li F, Liu F, Cheng JP, Bo XC, Wang SQ, Zhou WX, Zhang YX. Neuroendocrine immunomodulation network dysfunction in SAMP8 mice and PrP-hAβPPswe/PS1ΔE9 mice: potential mechanism underlying cognitive impairment. Oncotarget 2018; 7:22988-3005. [PMID: 27049828 PMCID: PMC5029605 DOI: 10.18632/oncotarget.8453] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 03/18/2016] [Indexed: 12/29/2022] Open
Abstract
Senescence-accelerated mouse prone 8 strain (SAMP8) and PrP-hAβPPswe/PS1ΔE9 (APP/PS1) mice are classic animal models of sporadic Alzheimer's disease and familial AD respectively. Our study showed that object recognition memory, spatial learning and memory, active and passive avoidance were deteriorated and neuroendocrine immunomodulation (NIM) network was imbalance in SAMP8 and APP/PS1 mice. SAMP8 and APP/PS1 mice had their own specific phenotype of cognition, neuroendocrine, immune and NIM molecular network. The endocrine hormone corticosterone, luteinizing hormone and follicle-stimulating hormone, chemotactic factor monocyte chemotactic protein-1, macrophage inflammatory protein-1β, regulated upon activation normal T cell expressed and secreted factor and eotaxin, pro-inflammatory factor interleukin-23, and the Th1 cell acting as cell immunity accounted for cognitive deficiencies in SAMP8 mice, while adrenocorticotropic hormone and gonadotropin-releasing hormone, colony stimulating factor granulocyte colony stimulating factor, and Th2 cell acting as humoral immunity in APP/PS1 mice. On the pathway level, chemokine signaling and T cell receptor signaling pathway played the key role in cognition impairments of two models, while cytokine-cytokine receptor interaction and natural killer cell mediated cytotoxicity were more important in cognitive deterioration of SAMP8 mice than APP/PS1 mice. This mechanisms of NIM network underlying cognitive impairment is significant for further understanding the pathogenesis of AD and can provide useful information for development of AD therapeutic drug.
Collapse
Affiliation(s)
- Jian-Hui Wang
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Xiao-Rui Cheng
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Xiao-Rui Zhang
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Tong-Xing Wang
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Wen-Jian Xu
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Fei Li
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Feng Liu
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Jun-Ping Cheng
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Xiao-Chen Bo
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Sheng-Qi Wang
- Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Wen-Xia Zhou
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Yong-Xiang Zhang
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| |
Collapse
|
248
|
Lecocq S, Spinella K, Dubois B, Lista S, Hampel H, Penner G. Aptamers as biomarkers for neurological disorders. Proof of concept in transgenic mice. PLoS One 2018; 13:e0190212. [PMID: 29304088 PMCID: PMC5755763 DOI: 10.1371/journal.pone.0190212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 12/11/2017] [Indexed: 11/18/2022] Open
Abstract
The act of selecting aptamers against blood serum leads to deep libraries of oligonucleotide sequences that bind to a range of epitopes in blood. In this study we developed an enriched aptamer library by performing positive selection against a pool of blood serum samples from transgenic mice (P301S) carrying the human tau gene and counter selecting against pooled blood serum from negative segregant (wild type) mice. We demonstrated that a large proportion of the aptamer sequences observed with next generation sequence (NGS) analysis were the same from selection round 5 and selection round 6. As a second step, we applied aliquots of the selection round 5 enriched library to blood serum from 16 individual mice for a single round of selection. Each of these individual libraries were characterized through NGS analysis and the changes in relative frequency in aptamers from transgenic mice versus wild type were used to construct a diagnostic fingerprint of the effect of the action of the transgene on the composition of blood serum. This study serves as a model for similar applications with human subjects.
Collapse
Affiliation(s)
| | | | - Bruno Dubois
- Centre des Maladies Cognitives et Comportementales (IM2A), Pavillon François Lhermitte, Hôpital University of Pitié Salpêtrière, Paris France
- Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié Salpêtrière, Paris, France
- UMR-S975, École des Neurosciences, Paris, France
- Université Pierre et Marie Curie-Paris 6, Paris, France
- AP-HP, Paris, France
| | - Simone Lista
- Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié Salpêtrière, Paris, France
- Université Pierre et Marie Curie-Paris 6, Paris, France
- AXA Research Fund & UPMC Chair, Sorbonne Universités, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Paris, France
| | - Harald Hampel
- Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié Salpêtrière, Paris, France
- Université Pierre et Marie Curie-Paris 6, Paris, France
- AXA Research Fund & UPMC Chair, Sorbonne Universités, Paris, France
- Inserm, Paris, France
- CNRS, Paris, France
- Département de Neurologie, Institut de la Mémoire et de la Maladie d’Alzheimer (IM2A), Hôpital Pitié-Salpêtrière, Paris, France
| | | |
Collapse
|
249
|
Tan CH, Fan CC, Mormino EC, Sugrue LP, Broce IJ, Hess CP, Dillon WP, Bonham LW, Yokoyama JS, Karch CM, Brewer JB, Rabinovici GD, Miller BL, Schellenberg GD, Kauppi K, Feldman HA, Holland D, McEvoy LK, Hyman BT, Bennett DA, Andreassen OA, Dale AM, Desikan RS. Polygenic hazard score: an enrichment marker for Alzheimer's associated amyloid and tau deposition. Acta Neuropathol 2018; 135:85-93. [PMID: 29177679 PMCID: PMC5758038 DOI: 10.1007/s00401-017-1789-4] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/10/2017] [Accepted: 11/10/2017] [Indexed: 01/19/2023]
Abstract
There is an urgent need for identifying nondemented individuals at the highest risk of progressing to Alzheimer's disease (AD) dementia. Here, we evaluated whether a recently validated polygenic hazard score (PHS) can be integrated with known in vivo cerebrospinal fluid (CSF) or positron emission tomography (PET) biomarkers of amyloid, and CSF tau pathology to prospectively predict cognitive and clinical decline in 347 cognitive normal (CN; baseline age range = 59.7-90.1, 98.85% white) and 599 mild cognitively impaired (MCI; baseline age range = 54.4-91.4, 98.83% white) individuals from the Alzheimer's Disease Neuroimaging Initiative 1, GO, and 2. We further investigated the association of PHS with post-mortem amyloid load and neurofibrillary tangles in the Religious Orders Study and Memory and Aging Project (ROSMAP) cohort (N = 485, age at death range = 71.3-108.3). In CN and MCI individuals, we found that amyloid and total tau positivity systematically varies as a function of PHS. For individuals in greater than the 50th percentile PHS, the positive predictive value for amyloid approached 100%; for individuals in less than the 25th percentile PHS, the negative predictive value for total tau approached 85%. High PHS individuals with amyloid and tau pathology showed the steepest longitudinal cognitive and clinical decline, even among APOE ε4 noncarriers. Among the CN subgroup, we similarly found that PHS was strongly associated with amyloid positivity and the combination of PHS and biomarker status significantly predicted longitudinal clinical progression. In the ROSMAP cohort, higher PHS was associated with higher post-mortem amyloid load and neurofibrillary tangles, even in APOE ε4 noncarriers. Together, our results show that even after accounting for APOE ε4 effects, PHS may be useful in MCI and preclinical AD therapeutic trials to enrich for biomarker-positive individuals at highest risk for short-term clinical progression.
Collapse
Affiliation(s)
- Chin Hong Tan
- Neuroradiology Section, Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA.
| | - Chun Chieh Fan
- Department of Cognitive Science, University of California, La Jolla, San Diego, CA, USA
| | - Elizabeth C Mormino
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Leo P Sugrue
- Neuroradiology Section, Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Iris J Broce
- Neuroradiology Section, Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Christopher P Hess
- Neuroradiology Section, Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - William P Dillon
- Neuroradiology Section, Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Luke W Bonham
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Jennifer S Yokoyama
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Celeste M Karch
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - James B Brewer
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
- Department of Radiology, University of California, La Jolla, San Diego, CA, USA
- Shiley-Marcos Alzheimer's Disease Research Center, University of California, La Jolla, San Diego, CA, USA
| | - Gil D Rabinovici
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce L Miller
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Karolina Kauppi
- Department of Radiology, University of California, La Jolla, San Diego, CA, USA
| | - Howard A Feldman
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
| | - Dominic Holland
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
| | - Linda K McEvoy
- Department of Radiology, University of California, La Jolla, San Diego, CA, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Ole A Andreassen
- NORMENT Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Anders M Dale
- Department of Cognitive Science, University of California, La Jolla, San Diego, CA, USA
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
- Department of Radiology, University of California, La Jolla, San Diego, CA, USA
| | - Rahul S Desikan
- Neuroradiology Section, Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA.
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
250
|
Chen M, Du ZY, Zheng X, Li DL, Zhou RP, Zhang K. Use of curcumin in diagnosis, prevention, and treatment of Alzheimer's disease. Neural Regen Res 2018; 13:742-752. [PMID: 29722330 PMCID: PMC5950688 DOI: 10.4103/1673-5374.230303] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
This review summarizes and describes the use of curcumin in diagnosis, prevention, and treatment of Alzheimer's disease. For diagnosis of Alzheimer's disease, amyloid-β and highly phosphorylated tau protein are the major biomarkers. Curcumin was developed as an early diagnostic probe based on its natural fluorescence and high binding affinity to amyloid-β. Because of its multi-target effects, curcumin has protective and preventive effects on many chronic diseases such as cerebrovascular disease, hypertension, and hyperlipidemia. For prevention and treatment of Alzheimer's disease, curcumin has been shown to effectively maintain the normal structure and function of cerebral vessels, mitochondria, and synapses, reduce risk factors for a variety of chronic diseases, and decrease the risk of Alzheimer's disease. The effect of curcumin on Alzheimer's disease involves multiple signaling pathways: anti-amyloid and metal iron chelating properties, antioxidation and anti-inflammatory activities. Indeed, there is a scientific basis for the rational application of curcumin in prevention and treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Min Chen
- Institute of Natural Medicinal Chemistry & Green Chemistry, College of Light Industry and Chemical Engineering, Guangdong University of Technology, Guangzhou, Guangdong Province, China
| | - Zhi-Yun Du
- Institute of Natural Medicinal Chemistry & Green Chemistry, College of Light Industry and Chemical Engineering, Guangdong University of Technology, Guangzhou, Guangdong Province, China
| | - Xi Zheng
- Institute of Natural Medicinal Chemistry & Green Chemistry, College of Light Industry and Chemical Engineering, Guangdong University of Technology, Guangzhou, Guangdong Province, China; Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA; Wuyi University; International Healthcare Innovation Institute (Jiangmen), Jiangmen, Guangdong Province, China
| | - Dong-Li Li
- Wuyi University; International Healthcare Innovation Institute (Jiangmen), Jiangmen, Guangdong Province, China
| | - Ren-Ping Zhou
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Kun Zhang
- Institute of Natural Medicinal Chemistry & Green Chemistry, College of Light Industry and Chemical Engineering, Guangdong University of Technology, Guangzhou; Wuyi University, Jiangmen, Guangdong Province, China; International Healthcare Innovation Institute (Jiangmen), Jiangmen, Guangdong Province, China
| |
Collapse
|