1
|
Zhang Q, Yang G, Luo Y, Jiang L, Chi H, Tian G. Neuroinflammation in Alzheimer's disease: insights from peripheral immune cells. Immun Ageing 2024; 21:38. [PMID: 38877498 PMCID: PMC11177389 DOI: 10.1186/s12979-024-00445-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024]
Abstract
Alzheimer's disease (AD) is a serious brain disorder characterized by the presence of beta-amyloid plaques, tau pathology, inflammation, neurodegeneration, and cerebrovascular dysfunction. The presence of chronic neuroinflammation, breaches in the blood-brain barrier (BBB), and increased levels of inflammatory mediators are central to the pathogenesis of AD. These factors promote the penetration of immune cells into the brain, potentially exacerbating clinical symptoms and neuronal death in AD patients. While microglia, the resident immune cells of the central nervous system (CNS), play a crucial role in AD, recent evidence suggests the infiltration of cerebral vessels and parenchyma by peripheral immune cells, including neutrophils, T lymphocytes, B lymphocytes, NK cells, and monocytes in AD. These cells participate in the regulation of immunity and inflammation, which is expected to play a huge role in future immunotherapy. Given the crucial role of peripheral immune cells in AD, this article seeks to offer a comprehensive overview of their contributions to neuroinflammation in the disease. Understanding the role of these cells in the neuroinflammatory response is vital for developing new diagnostic markers and therapeutic targets to enhance the diagnosis and treatment of AD patients.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Laboratory Medicine, Southwest Medical University, Luzhou, China
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH, USA
| | - Yuan Luo
- Department of Laboratory Medicine, Southwest Medical University, Luzhou, China
| | - Lai Jiang
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, China.
| | - Gang Tian
- Department of Laboratory Medicine, Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, The Affiliated Hospital of Southwest Medical University, Sichuan, 646000, China.
| |
Collapse
|
2
|
Cui W, Lv C, Geng P, Fu M, Zhou W, Xiong M, Li T. Novel targets and therapies of metformin in dementia: old drug, new insights. Front Pharmacol 2024; 15:1415740. [PMID: 38881878 PMCID: PMC11176471 DOI: 10.3389/fphar.2024.1415740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 05/16/2024] [Indexed: 06/18/2024] Open
Abstract
Dementia is a devastating disorder characterized by progressive and persistent cognitive decline, imposing a heavy public health burden on the individual and society. Despite numerous efforts by researchers in the field of dementia, pharmacological treatments are limited to relieving symptoms and fail to prevent disease progression. Therefore, studies exploring novel therapeutics or repurposing classical drugs indicated for other diseases are urgently needed. Metformin, a first-line antihyperglycemic drug used to treat type 2 diabetes, has been shown to be beneficial in neurodegenerative diseases including dementia. This review discusses and evaluates the neuroprotective role of metformin in dementia, from the perspective of basic and clinical studies. Mechanistically, metformin has been shown to improve insulin resistance, reduce neuronal apoptosis, and decrease oxidative stress and neuroinflammation in the brain. Collectively, the current data presented here support the future potential of metformin as a potential therapeutic strategy for dementia. This study also inspires a new field for future translational studies and clinical research to discover novel therapeutic targets for dementia.
Collapse
Affiliation(s)
- Wenxing Cui
- College of Life Sciences, Northwest University, Xi'an, China
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Chen Lv
- Hangzhou Simo Co., Ltd., Hangzhou, China
| | - Panling Geng
- College of Life Sciences, Northwest University, Xi'an, China
| | - Mingdi Fu
- College of Life Sciences, Northwest University, Xi'an, China
| | - Wenjing Zhou
- College of Life Sciences, Northwest University, Xi'an, China
| | - Mingxiang Xiong
- College of Life Sciences, Northwest University, Xi'an, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
3
|
Peng H, Mao C, Zhang J, Wang F, Jiang H, Zhang Y, Zhu Z, Zhong C, Xu F, Liu CF, Zhang Y. Cohort Profile: Heart and Brain Investigation in Taicang (HABIT) study. Int J Epidemiol 2024; 53:dyad192. [PMID: 38205861 DOI: 10.1093/ije/dyad192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Affiliation(s)
- Hao Peng
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, China
| | - Chengjie Mao
- Department of Neurology and Clinical Research Center of Neurological Disease, Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianan Zhang
- Department of Chronic Disease, Taicang Center of Disease Prevention and Control, Suzhou, China
| | - Fenchun Wang
- Department of Chronic Disease, Taicang Center of Disease Prevention and Control, Suzhou, China
| | - Hai Jiang
- Department of Chronic Disease, Taicang Center of Disease Prevention and Control, Suzhou, China
| | - Yingchun Zhang
- Department of Ultrasound, Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhengbao Zhu
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, China
| | - Chongke Zhong
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, China
| | - Feng Xu
- Department of Chronic Disease, Taicang Center of Disease Prevention and Control, Suzhou, China
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, Second Affiliated Hospital of Soochow University, Suzhou, China
- Institutes of Neuroscience, Soochow University, Suzhou, China
| | - Yonghong Zhang
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, China
| |
Collapse
|
4
|
Deng Z, Xie D, Cai J, Jiang J, Pan D, Liao H, Liu X, Xu Y, Li H, Shen Q, Lattanzi S, Xiao S, Tang Y. Different types of milk consumption and the risk of dementia: Analysis from a large-scale cohort study. Clin Nutr 2023; 42:2058-2067. [PMID: 37677911 DOI: 10.1016/j.clnu.2023.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 08/16/2023] [Accepted: 08/28/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND & AIMS Previous studies have investigated whether milk consumption has a role in preventing the development of cognitive impairment, but the results were inconsistent. Importantly, most of them have disregarded the role of different types of milk. This study aimed to examine the associations between different types of milk consumption and the risk of dementia. METHODS In this large-scale cohort study, participants without cognitive impairment at baseline were included from the UK Biobank. The type of milk mainly used was self-reported at baseline, including full-cream milk, skimmed-milk, soy milk, other milk, and no milk. The primary outcome was all-cause dementia. Secondary outcomes included Alzheimer's disease and vascular dementia. RESULTS Of the 307,271 participants included in the study (mean age 56.3 [SD 8.1] years), 3789 (1.2%) incident all-cause dementia cases were observed over a median follow-up of 12.3 years. After adjustment for potential confounders, only soy milk consumers had a statistically significantly lower risk of all-cause dementia compared with no milk consumers (hazard ratio [HR], 0.69; 95% confidence interval [CI], 0.54 to 0.90). When compared with soy milk non-consumers consisting of full-cream milk, skimmed-milk, and other milk consumers, soy milk consumers still showed a lower risk of all-cause dementia (HR, 0.76; 95% CI, 0.63 to 0.92), and there was no significant interaction with genetic risk for dementia (P for interaction = 0.15). Soy milk consumers showed a lower risk of Alzheimer's disease (HR, 0.70; 95% CI, 0.51 to 0.94; P = 0.02), while the association was not significant for vascular dementia (HR, 0.72; 95% CI, 0.47 to 1.12; P = 0.14). CONCLUSIONS The main consumption of soy milk was associated with a lower risk of dementia, particularly non-vascular dementia. Additional studies are needed to investigate how this association varies with the dose or frequency of the consumption of soy milk and to examine the generalizability of these findings in different populations.
Collapse
Affiliation(s)
- Zhenhong Deng
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Dongshu Xie
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jinhua Cai
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jingru Jiang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Dong Pan
- Department of Neurology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Huanquan Liao
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xingyi Liu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yongteng Xu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Honghong Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Qingyu Shen
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Simona Lattanzi
- Neurological Clinic, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Songhua Xiao
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China.
| | - Yamei Tang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
5
|
Thompson F, Russell S, Quigley R, Sagigi B, Miller G, Esterman A, Harriss LR, Taylor S, McDermott R, Strivens E. Dementia Risk Models in an Australian First Nations Population: Cross-Sectional Associations and Preparation for Follow-Up. J Alzheimers Dis Rep 2023; 7:543-555. [PMID: 37313487 PMCID: PMC10259055 DOI: 10.3233/adr-220093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 05/01/2023] [Indexed: 06/15/2023] Open
Abstract
Background Reducing the burden of dementia in First Nations populations may be addressed through developing population specific methods to quantify future risk of dementia. Objective To adapt existing dementia risk models to cross-sectional dementia prevalence data from a First Nations population in the Torres Strait region of Australia in preparation for follow-up of participants. To explore the diagnostic utility of these dementia risk models at detecting dementia. Methods A literature review to identify existing externally validated dementia risk models. Adapting these models to cross-sectional data and assessing their diagnostic utility through area under the receiver operating characteristic curve (AUROC) analyses and calibration using Hosmer-Lemeshow Chi2. Results Seven risk models could be adapted to the study data. The Aging, Cognition and Dementia (AgeCoDe) study, the Framingham Heart Study (FHS), and the Brief Dementia Screening Indicator (BDSI) had moderate diagnostic utility in identifying dementia (i.e., AUROC >0.70) before and after points for older age were removed. Conclusion Seven existing dementia risk models could be adapted to this First Nations population, and three had some cross-sectional diagnostic utility. These models were designed to predict dementia incidence, so their applicability to identify prevalent cases would be limited. The risk scores derived in this study may have prognostic utility as participants are followed up over time. In the interim, this study highlights considerations when transporting and developing dementia risk models for First Nations populations.
Collapse
Affiliation(s)
- Fintan Thompson
- Australian Institute of Tropical Health and Medicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Cairns, QLD, Australia
- Queensland Health, Cairns and Hinterland Hospital and Health Service, Cairns, QLD, Australia
- University of South Australia, SA, Australia
| | - Sarah Russell
- Queensland Health, Cairns and Hinterland Hospital and Health Service, Cairns, QLD, Australia
- College of Medicine and Dentistry, James Cook University, Cairns, QLD, Australia
| | - Rachel Quigley
- Queensland Health, Cairns and Hinterland Hospital and Health Service, Cairns, QLD, Australia
- College of Medicine and Dentistry, James Cook University, Cairns, QLD, Australia
| | - Betty Sagigi
- Queensland Health, Torres and Cape Hospital and Health Service, Thursday Island, QLD, Australia
| | - Gavin Miller
- Queensland Health, Cairns and Hinterland Hospital and Health Service, Cairns, QLD, Australia
| | | | - Linton R. Harriss
- Australian Institute of Tropical Health and Medicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Cairns, QLD, Australia
- Queensland Health, Cairns and Hinterland Hospital and Health Service, Cairns, QLD, Australia
| | - Sean Taylor
- Top End Health Service, Northern Territory Government, Darwin, NT, Australia
| | | | - Edward Strivens
- Queensland Health, Cairns and Hinterland Hospital and Health Service, Cairns, QLD, Australia
- College of Medicine and Dentistry, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
6
|
Lee AJ, Sanchez D, Reyes-Dumeyer D, Brickman AM, Lantigua RA, Vardarajan BN, Mayeux R. Reliability and Validity of self-reported Vascular Risk Factors in a Multi-Ethnic Community Based Study of Aging and Dementia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.12.23288492. [PMID: 37131736 PMCID: PMC10153321 DOI: 10.1101/2023.04.12.23288492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
INTRODUCTION The reliability and validity of self-reported cardiovascular and cerebrovascular risk factors remains inconsistent in aging research. METHODS We assessed the reliability, validity, sensitivity, specificity, and percent agreement of self-reported hypertension, diabetes, and heart disease, in comparison with direct measures of blood pressure, hemoglobin A1c (HbA1c), and medication use in 1870 participants in a multiethic study of aging and dementia. RESULTS Reliability of self-reported for hypertension, diabetes, and heart disease was excellent. Agreement between self-reports and clinical measures was moderate for hypertension (kappa: 0.58), good for diabetes (kappa: 0.76-0.79), and moderate for heart disease (kappa: 0.45) differing slightly by age, sex, education, and race/ethnic group. Sensitivity and specificity for hypertension was 88.6%-78.1%, for diabetes was 87.7%-92.0% (HbA1c > 6.5%) or 92.7%-92.8% (HbA1c > 7%), and for heart disease was 85.8%-75.5%. DISCUSSION Self-reported history of hypertension, diabetes, and heart disease are reliable and valid compared to direct measurements or medication use.
Collapse
|
7
|
Lee AJ, Sanchez D, Reyes-Dumeyer D, Brickman AM, Lantigua RA, Vardarajan BN, Mayeux R. Reliability and Validity of Self-Reported Vascular Risk Factors: Hypertension, Diabetes, and Heart Disease, in a Multi-Ethnic Community Based Study of Aging and Dementia. J Alzheimers Dis 2023; 95:275-285. [PMID: 37483004 PMCID: PMC10578288 DOI: 10.3233/jad-230374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2023] [Indexed: 07/25/2023]
Abstract
BACKGROUND Queries for the presence of cardiovascular and cerebrovascular risk factors are typically assessed through self-report. However, the reliability and validity of self-reported cardiovascular and cerebrovascular risk factors remain inconsistent in aging research. OBJECTIVE To determine the reliability and validity of the most frequently self-reported vascular risk factors: hypertension, diabetes, and heart disease. METHODS 1,870 individuals aged 65 years or older among African Americans, Caribbean Hispanics, and white non-Hispanic individuals were recruited as part of a community study of aging and dementia. We assessed the reliability, validity, sensitivity, specificity, and percent agreement of self-reported hypertension, diabetes, and heart disease, in comparison with direct measures of blood pressure, hemoglobin A1c (HbA1c), and medication use. The analyses were subsequently stratified by age, sex, education, and ethnic group. RESULTS Reliability of self-reported hypertension, diabetes, and heart disease was excellent. Agreement between self-reports and clinical measures was moderate for hypertension (kappa: 0.58), good for diabetes (kappa: 0.76-0.79), and moderate for heart disease (kappa: 0.45) differing slightly by age, sex, education, and ethnic group. Sensitivity and specificity for hypertension was 88.6% -78.1%, for diabetes was 87.7% -92.0% (HbA1c ≥6.5%) or 92.7% -92.8% (HbA1c ≥7%), and for heart disease was 85.8% -75.5%. Percent agreement of self-reported was 87.0% for hypertension, 91.6% -92.6% for diabetes, and 77.4% for heart disease. CONCLUSION Ascertainment of self-reported histories of hypertension, diabetes, and heart disease are reliable and valid compared to direct measurements or medication use.
Collapse
Affiliation(s)
- Annie J. Lee
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, New York, NY, USA
| | - Didi Sanchez
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, New York, NY, USA
| | - Dolly Reyes-Dumeyer
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, New York, NY, USA
| | - Adam M. Brickman
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, New York, NY, USA
| | - Rafael A. Lantigua
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, New York, NY, USA
| | - Badri N. Vardarajan
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, New York, NY, USA
| | - Richard Mayeux
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, New York, NY, USA
- Department of Medicine, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, New York, NY, USA
| |
Collapse
|
8
|
Reyes‐Dumeyer D, Faber K, Vardarajan B, Goate A, Renton A, Chao M, Boeve B, Cruchaga C, Pericak‐Vance M, Haines JL, Rosenberg R, Tsuang D, Sweet RA, Bennett DA, Wilson RS, Foroud T, Mayeux R. The National Institute on Aging Late-Onset Alzheimer's Disease Family Based Study: A resource for genetic discovery. Alzheimers Dement 2022; 18:1889-1897. [PMID: 34978149 PMCID: PMC9250549 DOI: 10.1002/alz.12514] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/25/2021] [Accepted: 08/11/2021] [Indexed: 02/02/2023]
Abstract
INTRODUCTION The National Institute on Aging Late-Onset Alzheimer's Disease Family Based Study (NIA-LOAD FBS) was established to study the genetic etiology of Alzheimer's disease (AD). METHODS Recruitment focused on families with two living affected siblings and a third first-degree relative similar in age with or without dementia. Uniform assessments were completed, DNA was obtained, as was neuropathology, when possible. Apolipoprotein E (APOE) genotypes, genome-wide single nucleotide polymorphism (SNP) arrays, and sequencing was completed in most families. RESULTS APOE genotype modified the age-at-onset in many large families. Novel variants and known variants associated with early- and late-onset AD and frontotemporal dementia were identified supporting an international effort to solve AD genetics. DISCUSSION The NIA-LOAD FBS is the largest collection of familial AD worldwide, and data or samples have been included in 123 publications addressing the genetic etiology of AD. Genetic heterogeneity and variability in the age-at-onset provides opportunities to investigate the complexity of familial AD.
Collapse
Affiliation(s)
- Dolly Reyes‐Dumeyer
- Department of NeurologyTaub Institute for Research on Alzheimer's Disease and the Aging Brain and the Gertrude H. Sergievsky Center, Columbia University in the City of New YorkNew YorkNew YorkUSA
| | - Kelley Faber
- Department of Medical and Molecular GeneticsNational Centralized Repository for Alzheimer's Disease and Related Dementias (NCRAD)Indiana University School of MedicineIndianapolisIndianaUSA
| | - Badri Vardarajan
- Department of NeurologyTaub Institute for Research on Alzheimer's Disease and the Aging Brain and the Gertrude H. Sergievsky Center, Columbia University in the City of New YorkNew YorkNew YorkUSA
| | - Alison Goate
- Department of Genetics & Genomic SciencesRonald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Alan Renton
- Department of Genetics & Genomic SciencesRonald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Michael Chao
- Department of Genetics & Genomic SciencesRonald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Brad Boeve
- Department of NeurologyMayo ClinicRochesterMinnesotaUSA
| | - Carlos Cruchaga
- Department of PsychiatryWashington University in St. LouisSt. LouisMissouriUSA
| | - Margaret Pericak‐Vance
- John P. Hussman Institute for Human GenomicsDr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of MedicineMiamiFloridaUSA
| | - Jonathan L. Haines
- Department of Population & Quantitative Health Sciences and Cleveland Institute for Computational BiologyCase Western Reserve UniversityClevelandOhioUSA
| | - Roger Rosenberg
- Department of NeurologyUniversity of Texas Southwestern Medical Center at DallasDallasTexasUSA
| | - Debby Tsuang
- GRECC VA Puget SoundDepartment of Psychiatry and Behavioral SciencesUniversity of WashingtonSeattleWashingtonUSA
| | - Robert A. Sweet
- Departments of Psychiatry and NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - David A. Bennett
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | - Robert S. Wilson
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | - Tatiana Foroud
- Department of Medical and Molecular GeneticsNational Centralized Repository for Alzheimer's Disease and Related Dementias (NCRAD)Indiana University School of MedicineIndianapolisIndianaUSA
| | - Richard Mayeux
- Department of NeurologyTaub Institute for Research on Alzheimer's Disease and the Aging Brain and the Gertrude H. Sergievsky Center, Columbia University in the City of New YorkNew YorkNew YorkUSA
| |
Collapse
|
9
|
Lee AJ, Raghavan NS, Bhattarai P, Siddiqui T, Sariya S, Reyes-Dumeyer D, Flowers XE, Cardoso SAL, De Jager PL, Bennett DA, Schneider JA, Menon V, Wang Y, Lantigua RA, Medrano M, Rivera D, Jiménez-Velázquez IZ, Kukull WA, Brickman AM, Manly JJ, Tosto G, Kizil C, Vardarajan BN, Mayeux R. FMNL2 regulates gliovascular interactions and is associated with vascular risk factors and cerebrovascular pathology in Alzheimer's disease. Acta Neuropathol 2022; 144:59-79. [PMID: 35608697 PMCID: PMC9217776 DOI: 10.1007/s00401-022-02431-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) has been associated with cardiovascular and cerebrovascular risk factors (CVRFs) during middle age and later and is frequently accompanied by cerebrovascular pathology at death. An interaction between CVRFs and genetic variants might explain the pathogenesis. Genome-wide, gene by CVRF interaction analyses for AD, in 6568 patients and 8101 controls identified FMNL2 (p = 6.6 × 10-7). A significant increase in FMNL2 expression was observed in the brains of patients with brain infarcts and AD pathology and was associated with amyloid and phosphorylated tau deposition. FMNL2 was also prominent in astroglia in AD among those with cerebrovascular pathology. Amyloid toxicity in zebrafish increased fmnl2a expression in astroglia with detachment of astroglial end feet from blood vessels. Knockdown of fmnl2a prevented gliovascular remodeling, reduced microglial activity and enhanced amyloidosis. APP/PS1dE9 AD mice also displayed increased Fmnl2 expression and reduced the gliovascular contacts independent of the gliotic response. Based on this work, we propose that FMNL2 regulates pathology-dependent plasticity of the blood-brain-barrier by controlling gliovascular interactions and stimulating the clearance of extracellular aggregates. Therefore, in AD cerebrovascular risk factors promote cerebrovascular pathology which in turn, interacts with FMNL2 altering the normal astroglial-vascular mechanisms underlying the clearance of amyloid and tau increasing their deposition in brain.
Collapse
Affiliation(s)
- Annie J Lee
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, 710 West 168th Street, New York, NY, 10032, USA
| | - Neha S Raghavan
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, 710 West 168th Street, New York, NY, 10032, USA
| | - Prabesh Bhattarai
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, 710 West 168th Street, New York, NY, 10032, USA
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Tatzberg 41, 01307, Dresden, Germany
| | - Tohid Siddiqui
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Tatzberg 41, 01307, Dresden, Germany
| | - Sanjeev Sariya
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, 710 West 168th Street, New York, NY, 10032, USA
| | - Dolly Reyes-Dumeyer
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, 710 West 168th Street, New York, NY, 10032, USA
| | - Xena E Flowers
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
| | - Sarah A L Cardoso
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
| | - Philip L De Jager
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, 710 West 168th Street, New York, NY, 10032, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Vilas Menon
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, 710 West 168th Street, New York, NY, 10032, USA
| | - Yanling Wang
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Rafael A Lantigua
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- Department of Medicine, College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, 630 West 168th Street, New York, NY, 10032, USA
| | - Martin Medrano
- School of Medicine, Pontificia Universidad Catolica Madre y Maestra (PUCMM), Santiago, Dominican Republic
| | - Diones Rivera
- Department of Neurology, CEDIMAT, Plaza de la Salud, Santo Domingo, Dominican Republic
- School of Medicine, Universidad Pedro Henriquez Urena (UNPHU), Santo Domingo, Dominican Republic
| | - Ivonne Z Jiménez-Velázquez
- Department of Medicine, Medical Sciences Campus, University of Puerto Rico School of Medicine, San Juan, Puerto Rico, 00936, USA
| | - Walter A Kukull
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, 98195, USA
| | - Adam M Brickman
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, 710 West 168th Street, New York, NY, 10032, USA
| | - Jennifer J Manly
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, 710 West 168th Street, New York, NY, 10032, USA
| | - Giuseppe Tosto
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, 710 West 168th Street, New York, NY, 10032, USA
| | - Caghan Kizil
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, 710 West 168th Street, New York, NY, 10032, USA
- German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association, Tatzberg 41, 01307, Dresden, Germany
| | - Badri N Vardarajan
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, 710 West 168th Street, New York, NY, 10032, USA
| | - Richard Mayeux
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA.
- The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA.
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, 710 West 168th Street, New York, NY, 10032, USA.
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, 1051 Riverside Drive, New York, NY, 10032, USA.
| |
Collapse
|
10
|
Silva NCBS, Bracko O, Nelson AR, de Oliveira FF, Robison LS, Shaaban CE, Hainsworth AH, Price BR. Vascular cognitive impairment and dementia: An early career researcher perspective. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12310. [PMID: 35496373 PMCID: PMC9043906 DOI: 10.1002/dad2.12310] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/16/2022] [Accepted: 03/22/2022] [Indexed: 01/07/2023]
Abstract
The field of vascular contributions to cognitive impairment and dementia (VCID) is evolving rapidly. Research in VCID encompasses topics aiming to understand, prevent, and treat the detrimental effects of vascular disease burden in the human brain. In this perspective piece, early career researchers (ECRs) in the field provide an overview of VCID, discuss past and present efforts, and highlight priorities for future research. We emphasize the following critical points as the field progresses: (a) consolidate existing neuroimaging and fluid biomarkers, and establish their utility for pharmacological and non-pharmacological interventions; (b) develop new biomarkers, and new non-clinical models that better recapitulate vascular pathologies; (c) amplify access to emerging biomarker and imaging techniques; (d) validate findings from previous investigations in diverse populations, including those at higher risk of cognitive impairment (e.g., Black, Hispanic, and Indigenous populations); and (e) conduct randomized controlled trials within diverse populations with well-characterized vascular pathologies emphasizing clinically meaningful outcomes.
Collapse
Affiliation(s)
- Nárlon C. Boa Sorte Silva
- Djavad Mowafaghian Centre for Brain HealthDepartment of Physical TherapyFaculty of MedicineThe University of British ColumbiaVancouverBritish ColumbiaCanada
| | - Oliver Bracko
- Department of BiologyThe University of MiamiCoral GablesFloridaUSA
| | - Amy R. Nelson
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileAlabamaUSA
| | | | - Lisa S. Robison
- Department of Psychology and NeuroscienceNova Southeastern UniversityFort LauderdaleFloridaUSA
| | | | - Atticus H. Hainsworth
- Molecular & Clinical Sciences Research InstituteSt George's University of London, UKDepartment of NeurologySt George's University Hospitals NHS Foundation Trust LondonLondonUK
| | - Brittani R. Price
- Department of NeuroscienceTufts University School of MedicineBostonMassachusettsUSA
| |
Collapse
|
11
|
Athanasaki A, Melanis K, Tsantzali I, Stefanou MI, Ntymenou S, Paraskevas SG, Kalamatianos T, Boutati E, Lambadiari V, Voumvourakis KI, Stranjalis G, Giannopoulos S, Tsivgoulis G, Paraskevas GP. Type 2 Diabetes Mellitus as a Risk Factor for Alzheimer’s Disease: Review and Meta-Analysis. Biomedicines 2022; 10:biomedicines10040778. [PMID: 35453527 PMCID: PMC9029855 DOI: 10.3390/biomedicines10040778] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/17/2022] [Accepted: 03/25/2022] [Indexed: 01/16/2023] Open
Abstract
Alzheimer’s disease is the most common type of dementia, reaching 60–80% of case totals, and is one of the major global causes of the elderly population’s decline in functionality concerning daily life activities. Epidemiological research has already indicated that, in addition to several others metabolic factors, diabetes mellitus type 2 is a risk factor of Alzheimer’s disease. Many molecular pathways have been described, and at the same time, there are clues that suggest the connection between type 2 diabetes mellitus and Alzheimer’s disease, through specific genes, autophagy, and even inflammatory pathways. A systematic review with meta-analysis was conducted, and its main goal was to reveal the multilevel connection between these diseases.
Collapse
Affiliation(s)
- Athanasia Athanasaki
- Department of Neurology, Evangelismos Hospital, 10676 Athens, Greece; (A.A.); (S.N.)
| | - Konstantinos Melanis
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
| | - Ioanna Tsantzali
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
| | - Maria Ioanna Stefanou
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
| | - Sofia Ntymenou
- Department of Neurology, Evangelismos Hospital, 10676 Athens, Greece; (A.A.); (S.N.)
| | - Sotirios G. Paraskevas
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
| | - Theodosis Kalamatianos
- 1st Department of Neurosurgery, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 10676 Athens, Greece; (T.K.); (G.S.)
| | - Eleni Boutati
- 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (E.B.); (V.L.)
| | - Vaia Lambadiari
- 2nd Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (E.B.); (V.L.)
| | - Konstantinos I. Voumvourakis
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
| | - George Stranjalis
- 1st Department of Neurosurgery, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 10676 Athens, Greece; (T.K.); (G.S.)
| | - Sotirios Giannopoulos
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
| | - Georgios Tsivgoulis
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
| | - George P. Paraskevas
- 2nd Department of Neurology, School of Medicine, National and Kapodistrian University of Athens, “Attikon” University General Hospital, 12462 Athens, Greece; (K.M.); (I.T.); (M.I.S.); (S.G.P.); (K.I.V.); (S.G.); (G.T.)
- Correspondence: ; Tel.: +30-2105832466
| |
Collapse
|
12
|
Tsao CW, Aday AW, Almarzooq ZI, Alonso A, Beaton AZ, Bittencourt MS, Boehme AK, Buxton AE, Carson AP, Commodore-Mensah Y, Elkind MSV, Evenson KR, Eze-Nliam C, Ferguson JF, Generoso G, Ho JE, Kalani R, Khan SS, Kissela BM, Knutson KL, Levine DA, Lewis TT, Liu J, Loop MS, Ma J, Mussolino ME, Navaneethan SD, Perak AM, Poudel R, Rezk-Hanna M, Roth GA, Schroeder EB, Shah SH, Thacker EL, VanWagner LB, Virani SS, Voecks JH, Wang NY, Yaffe K, Martin SS. Heart Disease and Stroke Statistics-2022 Update: A Report From the American Heart Association. Circulation 2022; 145:e153-e639. [PMID: 35078371 DOI: 10.1161/cir.0000000000001052] [Citation(s) in RCA: 2700] [Impact Index Per Article: 1350.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND The American Heart Association, in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, diet, and weight) and health factors (cholesterol, blood pressure, and glucose control) that contribute to cardiovascular health. The Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure, valvular disease, venous disease, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The American Heart Association, through its Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States to provide the most current information available in the annual Statistical Update. The 2022 Statistical Update is the product of a full year's worth of effort by dedicated volunteer clinicians and scientists, committed government professionals, and American Heart Association staff members. This year's edition includes data on the monitoring and benefits of cardiovascular health in the population and an enhanced focus on social determinants of health, adverse pregnancy outcomes, vascular contributions to brain health, and the global burden of cardiovascular disease and healthy life expectancy. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
13
|
Rosen AC, Arias JJ, Ashford JW, Blacker D, Chhatwal JP, Chin NA, Clark L, Denny SS, Goldman JS, Gleason CE, Grill JD, Heidebrink JL, Henderson VW, Lavacot JA, Lingler JH, Menon M, Nosheny RL, Oliveira FF, Parker MW, Rahman-Filipiak A, Revoori A, Rumbaugh MC, Sanchez DL, Schindler SE, Schwarz CG, Toy L, Tyrone J, Walter S, Wang LS, Wijsman EM, Zallen DT, Aggarwal NT. The Advisory Group on Risk Evidence Education for Dementia: Multidisciplinary and Open to All. J Alzheimers Dis 2022; 90:953-962. [PMID: 35938255 PMCID: PMC9901285 DOI: 10.3233/jad-220458] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The brain changes of Alzheimer's disease and other degenerative dementias begin long before cognitive dysfunction develops, and in people with subtle cognitive complaints, clinicians often struggle to predict who will develop dementia. The public increasingly sees benefits to accessing dementia risk evidence (DRE) such as biomarkers, predictive algorithms, and genetic information, particularly as this information moves from research to demonstrated usefulness in guiding diagnosis and clinical management. For example, the knowledge that one has high levels of amyloid in the brain may lead one to seek amyloid reducing medications, plan for disability, or engage in health promoting behaviors to fight cognitive decline. Researchers often hesitate to share DRE data, either because they are insufficiently validated or reliable for use in individuals, or there are concerns about assuring responsible use and ensuring adequate understanding of potential problems when one's biomarker status is known. Concerns include warning people receiving DRE about situations in which they might be compelled to disclose their risk status potentially leading to discrimination or stigma. The Advisory Group on Risk Evidence Education for Dementia (AGREEDementia) welcomes all concerned with how best to share and use DRE. Supporting understanding in clinicians, stakeholders, and people with or at risk for dementia and clearly delineating risks, benefits, and gaps in knowledge is vital. This brief overview describes elements that made this group effective as a model for other health conditions where there is interest in unfettered collaboration to discuss diagnostic uncertainty and the appropriate use and communication of health-related risk information.
Collapse
Affiliation(s)
- Allyson C. Rosen
- VA Medical Center-Palo Alto, Palo Alto, CA, USA,Stanford University, School of Medicine, Stanford, CA, USA,Correspondence to: Allyson C. Rosen, PhD, ABPP-CN, Mental Illness Research, Education and Clinical Center (MIRECC), Palo Alto VA Medical Center, 3801 Miranda Ave (151Y), Palo Alto, CA 94304-1207, USA. Tel.: +1 650 279 3949;
| | - Jalayne J. Arias
- School of Public Health Georgia State University, Atlanta, GA, USA
| | - J. Wesson Ashford
- VA Medical Center-Palo Alto, Palo Alto, CA, USA,Stanford University, School of Medicine, Stanford, CA, USA
| | - Deborah Blacker
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Nathan A. Chin
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Lindsay Clark
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sharon S. Denny
- The Association for Frontotemporal Degeneration, King of Prussia, PA, USA
| | - Jill S. Goldman
- Columbia University Irving Medical Center, New York, NY, USA
| | - Carey E. Gleason
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | - Judith L. Heidebrink
- Michigan Alzheimer’s Disease Research Center, University of Michigan, Ann Arbor, MI, USA
| | - Victor W. Henderson
- Departments of Epidemiology & Population Health and of Neurology & Neurological Sciences, Stanford University, Stanford, CA, USA
| | | | | | | | - Rachel L. Nosheny
- Center for Imaging of Neurodegenerative Diseases, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | | | - Monica W. Parker
- Emory Goizueta Alzheimer’s Disease Research Center, Atlanta, GA, USA
| | | | | | | | | | - Suzanne E. Schindler
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA
| | | | - Leslie Toy
- VA Medical Center-Palo Alto, Palo Alto, CA, USA
| | - Jamie Tyrone
- Beating Alzheimer’s by Embracing Science, Ramona, CA, USA
| | - Sarah Walter
- Alzheimer’s Therapeutic Research Institute/USC, San Diego, CA, USA
| | - Li-san Wang
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | | | | | | |
Collapse
|
14
|
Rezaee N, Fernando WB, Hone E, Sohrabi HR, Johnson SK, Gunzburg S, Martins RN. Potential of Sorghum Polyphenols to Prevent and Treat Alzheimer's Disease: A Review Article. Front Aging Neurosci 2021; 13:729949. [PMID: 34690742 PMCID: PMC8527926 DOI: 10.3389/fnagi.2021.729949] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/24/2021] [Indexed: 12/06/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by the excessive deposition of extracellular amyloid-beta peptide (Aβ) and the build-up of intracellular neurofibrillary tangles containing hyperphosphorylated tau proteins. This leads to neuronal damage, cell death and consequently results in memory and learning impairments leading to dementia. Although the exact cause of AD is not yet clear, numerous studies indicate that oxidative stress, inflammation, and mitochondrial dysfunction significantly contribute to its onset and progression. There is no effective therapeutic approach to stop the progression of AD and its associated symptoms. Thus, early intervention, preferably, pre-clinically when the brain is not significantly affected, is a better option for effective treatment. Natural polyphenols (PP) target multiple AD-related pathways such as protecting the brain from Aβ and tau neurotoxicity, ameliorating oxidative damage and mitochondrial dysfunction. Among natural products, the cereal crop sorghum has some unique features. It is one of the major global grain crops but in the developed world, it is primarily used as feed for farm animals. A broad range of PP, including phenolic acids, flavonoids, and condensed tannins are present in sorghum grain including some classes such as proanthocyanidins that are rarely found in others plants. Pigmented varieties of sorghum have the highest polyphenolic content and antioxidant activity which potentially makes their consumption beneficial for human health through different pathways such as oxidative stress reduction and thus the prevention and treatment of neurodegenerative diseases. This review summarizes the potential of sorghum PP to beneficially affect the neuropathology of AD.
Collapse
Affiliation(s)
- Nasim Rezaee
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - W.M.A.D. Binosha Fernando
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Eugene Hone
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Hamid R. Sohrabi
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
- Centre for Healthy Ageing, Health Future Institute, Murdoch University, Murdoch, WA, Australia
| | - Stuart K. Johnson
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Perth, WA, Australia
- Ingredients by Design Pty Ltd., Lesmurdie, WA, Australia
| | | | - Ralph N. Martins
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
15
|
Wang H, Lu J, Zhao X, Qin R, Song K, Xu Y, Zhang J, Chen Y. Alzheimer's disease in elderly COVID-19 patients: potential mechanisms and preventive measures. Neurol Sci 2021; 42:4913-4920. [PMID: 34550494 PMCID: PMC8455804 DOI: 10.1007/s10072-021-05616-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 09/17/2021] [Indexed: 12/13/2022]
Abstract
Advanced age correlates with higher morbidity and mortality among patients affected with the novel coronavirus disease 2019 (COVID-19). Because systemic inflammation and neurological symptoms are also common in severe COVID-19 cases, there is concern that COVID-19 may lead to neurodegenerative conditions such as Alzheimer’s disease (AD). In this review, we summarize possible mechanisms by which infection with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19, may cause AD in elderly COVID-19 patients and describe preventive measures to mitigate risk. Potential mechanisms include NLRP3 inflammasome activation and IL-1β release, renin-angiotensin system hyperactivation, innate immune activation, oxidative stress, direct viral infection, and direct cytolytic β-cell damage. Anti-inflammatory therapies, including TNF-α inhibitors and nonsteroidal anti-inflammatory drugs, antioxidants such as the vitamin E family, nutritional intervention, physical activity, blood glucose control, and vaccination are proposed as preventive measures to minimize AD risk in COVID-19 patients. Since several risk factors for AD may converge during severe SARS-CoV-2 infection, neurologists should be alert for potential symptoms of AD and actively implement preventive measures in patients presenting with neuropsychiatric symptoms and in high-risk patients such as the elderly.
Collapse
Affiliation(s)
- Haili Wang
- Department of Clinical Medicine, Dalian Medical University, Dalian, 116000, Liaoning, China.,Department of Neurology, Clinical Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu, China
| | - Juan Lu
- Department of Neuro Intensive Care Unit, Subei People's Hospital of Jiangsu Province, Yangzhou, 225000, Jiangsu, China
| | - Xia Zhao
- Department of Emergency Medicine, Subei People's Hospital of Jiangsu Province, Yangzhou, 225000, Jiangsu, China
| | - Rongyin Qin
- Department of Neurology, Shanghai General Hospital (Jiading District), Jiading, Shanghai, 201812, China
| | - Kangping Song
- Department of Neurology, Institute of Clinical Neuroscience, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Yao Xu
- Department of Neurology, Clinical Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu, China
| | - Jun Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Jingan, Shanghai, 200040, China.
| | - Yingzhu Chen
- Department of Neurology, Clinical Medical College, Yangzhou University, Yangzhou, 225000, Jiangsu, China.
| |
Collapse
|
16
|
Kafadar AH, Barrett C, Cheung KL. Knowledge and perceptions of Alzheimer's disease in three ethnic groups of younger adults in the United Kingdom. BMC Public Health 2021; 21:1124. [PMID: 34118921 PMCID: PMC8196265 DOI: 10.1186/s12889-021-11231-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/27/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a global public health problem with an ageing population. Knowledge is essential to promote early awareness, diagnosis and treatment of AD symptoms. AD knowledge is influenced by many cultural factors including cultural beliefs, attitudes and language barriers. This study aims: (1) to define AD knowledge level and perceptions amongst adults between 18 and 49 years of age in the UK; (2) to compare knowledge and perceptions of AD among three main ethnic groups (Asian, Blacks, and Whites); and (3) to assess potential associations of age, gender, education level, affinity with older people (65 or over), family history and caregiving history with AD knowledge. METHODS Data was collected from 186 participants as a convenience sample of younger adults of three different ethnicities (16.1% Asian, 16.7% Black, 67.2% White), living in the UK, recruited via an online research platform. The majority of the participants were in the 18-34 years age group (87.6%). Demographic characteristics of participants and AD knowledge correlation were assessed by the 30-item Alzheimer's Disease Knowledge Scale (ADKS), comprising 7 content domains. ANOVA/ANCOVA were used to assess differences in AD knowledge by ethnicity, gender, education level, age and affinity with dementia and Alzheimer's patients. RESULTS For AD general knowledge across all respondents only 45.0% answers were correct. No significant differences were found for the total ADKS score between ethnicities in this younger age group, who did not differ in education level. However, there were significant knowledge differences for the ADKS symptom domain score even after controlling for other demographics variables such as gender, education level (p = 0.005). White respondents were more likely to know about AD symptoms than their Black counterparts (p = 0.026). CONCLUSION The study's findings suggest that the AD knowledge level is not adequate for all ethnic groups. Meanwhile, significant differences were observed in symptoms, between ethnic groups, and therefore, differ in their needs regards health communication. The study contributes to an understanding of ethnicity differences in AD knowledge amongst adults from 18 to 49 years of age in the UK and may also provide input into an intervention plan for different ethnicities' information needs.
Collapse
Affiliation(s)
| | - Christine Barrett
- Department of Health Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Kei Long Cheung
- Department of Health Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| |
Collapse
|
17
|
Chen Q, Wang L, Li C, Hu W, Fan Y, Chen Z, Wu L, Lu Z, Ye J, Chen S, Tong J, Ruan L, Mei J, Lu H. Chronic Cardio-Metabolic Disease Increases the Risk of Worse Outcomes Among Hospitalized Patients With COVID-19: A Multicenter, Retrospective, and Real-World Study. J Am Heart Assoc 2021; 10:e018451. [PMID: 34096317 PMCID: PMC8477891 DOI: 10.1161/jaha.120.018451] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background Although chronic cardio-metabolic disease is a common comorbidity among patients with COVID-19, its effects on the clinical characteristics and outcome are not well known. Methods and Results This study aimed to explore the association between underlying cardio-metabolic disease and mortality with COVID-19 among hospitalized patients. This multicenter, retrospective, and real-world study was conducted from January 22, 2020 to March 25, 2020 in China. Data between patients with and without 5 main cardio-metabolic diseases including hypertension, diabetes mellitus, coronary heart disease, cerebrovascular disease, and hyperlipidemia were compared. A total of 1303 hospitalized patients were included in the final analysis. Of them, 520 patients (39.9%) had cardio-metabolic disease. Compared with patients without cardio-metabolic disease, more patients with cardio-metabolic disease had COVID-related complications including acute respiratory distress syndrome (9.81% versus 3.32%; P<0.001), acute kidney injury (4.23% versus 1.40%; P=0.001), secondary infection (13.9% versus 9.8%; P=0.026), hypoproteinemia (12.1% versus 5.75%; P<0.001), and coagulopathy (19.4% versus 10.3%; P<0.001), had higher incidences of the severe type of COVID-19 (32.9% versus 16.7%; P<0.001), more were admitted to the intensive care unit (11.7% versus 7.92%; P=0.021), and required mechanical ventilation (9.8% versus 4.3%; P<0.001). When the number of the patients' cardio-metabolic diseases was 0, 1, and >2, the mortality was 4.2%, 11.1%, and 19.8%, respectively. The multivariable-adjusted hazard ratio of mortality among patients with cardio-metabolic disease was 1.80 (95% CI, 1.17-2.77). Conclusions Cardio-metabolic disease was a common condition among hospitalized patients with COVID-19, and it was associated with higher risks of in-hospital mortality.
Collapse
Affiliation(s)
- Qijian Chen
- Department of Emergency the Fifth Hospital in Wuhan Wuhan Hubei China
| | - Lingling Wang
- Department of Endocrinology & Metabolism the Fifth Affiliated Hospital of Sun Yat-sen University Zhuhai Guangdong China.,Department of Gerontology the Fifth Affiliated Hospital of Sun Yat-sen University Zhuhai Guangdong China
| | - Chang Li
- Department of Cardiology Hubei No. 3 People's Hospital of Jianghan University Wuhan Hubei China
| | - Weihua Hu
- Department of Respiratory Medicine the First Hospital of Jingzhou Clinical Medical College Yangtze University Jingzhou Hubei China
| | - Yameng Fan
- School of Health Sciences Wuhan University Wuhan Hubei China
| | - Zaishu Chen
- People's Hospital of Jiayu County Xianning Hubei China
| | - Longlong Wu
- People's Hospital of Nanzhang County Nanzhang Hubei China
| | - Zhanjin Lu
- Department of Endocrinology & Metabolism the Fifth Affiliated Hospital of Sun Yat-sen University Zhuhai Guangdong China
| | - Jianfang Ye
- Department of Endocrinology & Metabolism the Fifth Affiliated Hospital of Sun Yat-sen University Zhuhai Guangdong China
| | - Shiyan Chen
- Department of Endocrinology & Metabolism the Fifth Affiliated Hospital of Sun Yat-sen University Zhuhai Guangdong China
| | - Junlu Tong
- Department of Endocrinology & Metabolism the Fifth Affiliated Hospital of Sun Yat-sen University Zhuhai Guangdong China
| | - Liemin Ruan
- Central Laboratory Ningbo First Hospital of Zhejiang University Ningbo Zhejiang China
| | - Jin Mei
- Central Laboratory Ningbo First Hospital of Zhejiang University Ningbo Zhejiang China.,Anatomy Department Wenzhou Medical UniversityWenzhou University Town Ningbo Zhejiang China
| | - Hongyun Lu
- Department of Endocrinology & Metabolism Zhuhai Hospital Affiliated with Jinan UniversityZhuhai People's Hospital Zhuhai Guangdong China
| |
Collapse
|
18
|
Torres S, Alexander A, O'Bryant S, Medina LD. Cognition and the Predictive Utility of Three Risk Scores in an Ethnically Diverse Sample. J Alzheimers Dis 2021; 75:1049-1059. [PMID: 32390625 DOI: 10.3233/jad-191284] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Various factors, such as age, cardiovascular concerns, and lifestyle patterns, are associated with risk for cognitive decline and Alzheimer's disease (AD). Risk scores model predictive risk of developing a disease (e.g., dementia, stroke). Many of these scores have been primarily developed in largely non-Hispanic/Latino (non-H/L) White samples and little is known about their applicability in ethno-racially diverse populations. OBJECTIVE The primary aim was to examine the relationship between three established risk scores and cognitive performance. These relationships were compared across ethnic groups. METHODS We conducted a cross-sectional study with a multi-ethnic, rural-dwelling group of participants (Mage = 61.6±12.6 years, range: 40-96 years; 373F:168M; 39.7% H/L). The Cardiovascular Risk Factors, Aging and Dementia (CAIDE), Framingham Risk Score (FRS), and Washington Heights-Inwood Columbia Aging Project (WHICAP) score were calculated for each participant. RESULTS All three scores were significantly associated with cognition in both H/L and non-H/L groups. In H/Ls, cognition was predicted by FRS: β= -0.08, p = 0.022; CAIDE: β= -0.08, p < 0.001; and WHICAP: β= -0.04, p < 0.001. In non-H/Ls, cognition was predicted by FRS: β= -0.11, p < 0.001; CAIDE: β= -0.14, p < 0.001; and WHICAP: β= -0.08, p < 0.001. The strength of this relationship differed between groups for FRS [t(246) = -4.61, p < 0.001] and CAIDE [t(420) = -3.20, p = 0.001], but not for WHICAP [t(384) = -1.03, p = 0.30], which already includes ethnicity in its calculation. CONCLUSION These findings support the utility of these three risk scores in predicting cognition while underscoring the need to account for ethnicity. Moreover, our results highlight the importance of cardiovascular and other demographic factors in predicting cognitive outcomes.
Collapse
Affiliation(s)
- Stephanie Torres
- Department of Psychology, University of Houston, Houston, TX, USA
| | - Angel Alexander
- Department of Psychology, University of Houston, Houston, TX, USA
| | - Sid O'Bryant
- Department of Pharmacology & Neuroscience, University of Northern Texas Health Science Center, Fort Worth, TX, USA
| | - Luis D Medina
- Department of Psychology, University of Houston, Houston, TX, USA
| |
Collapse
|
19
|
Fang J, Pieper AA, Nussinov R, Lee G, Bekris L, Leverenz JB, Cummings J, Cheng F. Harnessing endophenotypes and network medicine for Alzheimer's drug repurposing. Med Res Rev 2020; 40:2386-2426. [PMID: 32656864 PMCID: PMC7561446 DOI: 10.1002/med.21709] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 06/23/2020] [Accepted: 06/27/2020] [Indexed: 12/16/2022]
Abstract
Following two decades of more than 400 clinical trials centered on the "one drug, one target, one disease" paradigm, there is still no effective disease-modifying therapy for Alzheimer's disease (AD). The inherent complexity of AD may challenge this reductionist strategy. Recent observations and advances in network medicine further indicate that AD likely shares common underlying mechanisms and intermediate pathophenotypes, or endophenotypes, with other diseases. In this review, we consider AD pathobiology, disease comorbidity, pleiotropy, and therapeutic development, and construct relevant endophenotype networks to guide future therapeutic development. Specifically, we discuss six main endophenotype hypotheses in AD: amyloidosis, tauopathy, neuroinflammation, mitochondrial dysfunction, vascular dysfunction, and lysosomal dysfunction. We further consider how this endophenotype network framework can provide advances in computational and experimental strategies for drug-repurposing and identification of new candidate therapeutic strategies for patients suffering from or at risk for AD. We highlight new opportunities for endophenotype-informed, drug discovery in AD, by exploiting multi-omics data. Integration of genomics, transcriptomics, radiomics, pharmacogenomics, and interactomics (protein-protein interactions) are essential for successful drug discovery. We describe experimental technologies for AD drug discovery including human induced pluripotent stem cells, transgenic mouse/rat models, and population-based retrospective case-control studies that may be integrated with multi-omics in a network medicine methodology. In summary, endophenotype-based network medicine methodologies will promote AD therapeutic development that will optimize the usefulness of available data and support deep phenotyping of the patient heterogeneity for personalized medicine in AD.
Collapse
Affiliation(s)
- Jiansong Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Andrew A Pieper
- Harrington Discovery Institute, University Hospital Case Medical Center; Department of Psychiatry, Case Western Reserve University, Geriatric Research Education and Clinical Centers, Louis Stokes Cleveland VAMC, Cleveland, OH 44106, USA
| | - Ruth Nussinov
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Garam Lee
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV 89106, USA
| | - Lynn Bekris
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| | - James B. Leverenz
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jeffrey Cummings
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV 89106, USA
- Department of Brain Health, School of Integrated Health Sciences, UNLV, Las Vegas, NV 89154, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| |
Collapse
|
20
|
Pan Y, Jing J, Cai X, Wang Y, Wang S, Meng X, Zeng C, Shi J, Ji J, Lin J, Lyu L, Zhang Z, Mei L, Li S, Li S, Zhu W, Li H, Wei T, Wang Y. PolyvasculaR Evaluation for Cognitive Impairment and vaScular Events (PRECISE)-a population-based prospective cohort study: rationale, design and baseline participant characteristics. Stroke Vasc Neurol 2020; 6:145-151. [PMID: 32863279 PMCID: PMC8005894 DOI: 10.1136/svn-2020-000411] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/21/2020] [Accepted: 07/30/2020] [Indexed: 11/04/2022] Open
Abstract
Background and purpose Cardiovascular diseases and dementia are two major diseases in the elderly. Atherosclerosis is associated with future vascular events and cognitive impairment. The PolyvasculaR Evaluation for Cognitive Impairment and vaScular Events (PRECISE) study is a population-based prospective cohort study with comprehensive evaluation of multiterritorial artery stenosis and plaque using advanced vascular imaging techniques and prospective collection of vascular events and cognitive assessments. Methods Between May 2017 and September 2019, the PRECISE study enrolled 3067 community-dwelling adults with ages between 50 and 75 years cluster sampled from six villages and four communities of Lishui city in China. Data are collected in face-to-face interviews at baseline, 2-year and 4-year follow-up visits. Brain MRI including high-resolution sequences for intracranial and carotidal arteries and CT angiography for thoracoabdominal arteries were performed at baseline and will be rescanned after 4 years. Cardiovascular/cerebrovascular events and cognitive assessment will be prospectively collected after the enrollment. Blood and urine samples were collected and biomarkers were tested at baseline. Results A total of 3067 subjects were enrolled, among which 53.5% were female with an average age of 61.2±6.7 years. Among them, 2.8%, 8.1%, 43.1% and 21.6% had a history of stroke, coronary heart diseases, hypertension and diabetes mellitus, respectively. Conclusions The PRECISE study is a population-based prospective cohort study with comprehensive evaluation of atherosclerotic stenosis and plaque using advanced vascular imaging techniques. Data from this cohort provide us an opportunity to precisely evaluate polyvascular atherosclerosis and its association with future vascular events and cognitive impairment. Trial registration number ClinicalTrials.gov Registry (NCT03178448).
Collapse
Affiliation(s)
- Yuesong Pan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jing Jing
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xueli Cai
- Department of Neurology, Lishui Hospital, Zhejiang University School of Medicine, Lishui, China
| | - Yilong Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Suying Wang
- Cerebrovascular Research Lab, Lishui Hospital, Zhejiang University School of Medicine, Lishui, China
| | - Xia Meng
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Chunlai Zeng
- Department of Cardiology, Lishui Hospital, Zhejiang University School of Medicine, Lishui, China
| | - Jiong Shi
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Lishui Hospital, Zhejiang University School of Medicine, Lishui, China
| | - Jinxi Lin
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Lingchun Lyu
- Department of Cardiology, Lishui Hospital, Zhejiang University School of Medicine, Lishui, China
| | - Zhe Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Lerong Mei
- Cerebrovascular Research Lab, Lishui Hospital, Zhejiang University School of Medicine, Lishui, China
| | - Shiping Li
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Shan Li
- Cerebrovascular Research Lab, Lishui Hospital, Zhejiang University School of Medicine, Lishui, China
| | - Wanlin Zhu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Hao Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Tiemin Wei
- Department of Cardiology, Lishui Hospital, Zhejiang University School of Medicine, Lishui, China
| | - Yongjun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China .,China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
21
|
Effect of Combined Antihypertensive and Lipid-Lowering Therapies on Cognitive Function: A New Treatment Strategy? Cardiol Res Pract 2020; 2020:1484357. [PMID: 32351732 PMCID: PMC7178519 DOI: 10.1155/2020/1484357] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/03/2020] [Accepted: 03/19/2020] [Indexed: 01/06/2023] Open
Abstract
Risk factors for cardiovascular disease such as hypertension and hyperlipidemia are associated with cognitive decline. However, there is still no clear evidence that the use of antihypertensive or lipid-lowering therapy can prevent or delay cognitive decline or development of dementia. To provide a reference for clinical treatment, we analyzed the potential mechanisms of cognitive dysfunction induced by hypertension and hyperlipidemia, the clinical research and controversy of antihypertensive and lipid-lowering therapies on cognitive function, and the clinical value of combined antihypertensive and lipid-lowering therapy. It is currently believed that hypertension and elevated blood cholesterol levels in middle-aged people may be related to cognitive impairment or dementia in the elderly. Some studies suggest that intensive antihypertensive or lipid-lowering therapies are better than standard antihypertensive or lipid-lowering therapy, yet further tests are needed to confirm their effects on cognitive function. Actively controlling potential risk factors from middle age may be important for Alzheimer's disease (AD) prevention.
Collapse
|
22
|
Adiponectin and Cognitive Decline. Int J Mol Sci 2020; 21:ijms21062010. [PMID: 32188008 PMCID: PMC7139651 DOI: 10.3390/ijms21062010] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/10/2020] [Accepted: 03/13/2020] [Indexed: 12/11/2022] Open
Abstract
Adiponectin (ADPN) is a plasma protein secreted by adipose tissue showing pleiotropic effects with anti-diabetic, anti-atherogenic, and anti-inflammatory properties. Initially, it was thought that the main role was only the metabolism control. Later, ADPN receptors were also found in the central nervous system (CNS). In fact, the receptors AdipoR1 and AdipoR2 are expressed in various areas of the brain, including the hypothalamus, hippocampus, and cortex. While AdipoR1 regulates insulin sensitivity through the activation of the AMP-activated protein kinase (AMPK) pathway, AdipoR2 stimulates the neural plasticity through the activation of the peroxisome proliferator-activated receptor alpha (PPARα) pathway that inhibits inflammation and oxidative stress. Overall, based on its central and peripheral actions, ADPN appears to have neuroprotective effects by reducing inflammatory markers, such as C-reactive protein (PCR), interleukin 6 (IL6), and Tumor Necrosis Factor a (TNFa). Conversely, high levels of inflammatory cascade factors appear to inhibit the production of ADPN, suggesting bidirectional modulation. In addition, ADPN appears to have insulin-sensitizing action. It is known that a reduction in insulin signaling is associated with cognitive impairment. Based on this, it is of great interest to investigate the mechanism of restoration of the insulin signal in the brain as an action of ADPN, because it is useful for testing a possible pharmacological treatment for the improvement of cognitive decline. Anyway, if ADPN regulates neuronal functioning and cognitive performances by the glycemic metabolic system remains poorly explored. Moreover, although the mechanism is still unclear, women compared to men have a doubled risk of developing cognitive decline. Several studies have also supported that during the menopausal transition, the estrogen reduction can adversely affect the brain, in particular, verbal memory and verbal fluency. During the postmenopausal period, in obese and insulin-resistant individuals, ADPN serum levels are significantly reduced. Our recent study has evaluated the relationship between plasma ADPN levels and cognitive performances in menopausal women. Thus, the aim of this review is to summarize both the mechanisms and the effects of ADPN in the central nervous system and the relationship between plasma ADPN levels and cognitive performances, also in menopausal women.
Collapse
|
23
|
Chen R, Shu Y, Zeng Y. Links Between Adiponectin and Dementia: From Risk Factors to Pathophysiology. Front Aging Neurosci 2020; 11:356. [PMID: 31969813 PMCID: PMC6960116 DOI: 10.3389/fnagi.2019.00356] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 12/05/2019] [Indexed: 12/12/2022] Open
Abstract
With the aging population, dementia is becoming one of the most serious and troublesome global public health issues. Numerous studies have been seeking for effective strategies to delay or block its progression, but with little success. In recent years, adiponectin (APN) as one of the most abundant and multifunctional adipocytokines related to anti-inflammation, regulating glycogen metabolism and inhibiting insulin resistance (IR) and anti-atherosclerosis, has attracted widespread attention. In this article, we summarize recent studies that have contributed to a better understanding of the extent to which APN influences the risks of developing dementia as well as its pathophysiological progression. In addition, some controversial results interlinked with its effects on cognitive dysfunction diseases will be critically discussed. Ultimately, we aim to gain a novel insight into the pleiotropic effects of APN levels in circulation and suggest potential therapeutic target and future research strategies.
Collapse
Affiliation(s)
- RuiJuan Chen
- Department of Geriatrics, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi Shu
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi Zeng
- Department of Geriatrics, Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
24
|
Rasmussen KL, Tybjærg-Hansen A, Nordestgaard BG, Frikke-Schmidt R. Absolute 10-year risk of dementia by age, sex and APOE genotype: a population-based cohort study. CMAJ 2019; 190:E1033-E1041. [PMID: 30181149 DOI: 10.1503/cmaj.180066] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Dementia is a major cause of disability, and risk-factor reduction may have the potential to delay or prevent the disease. Our aim was to determine the absolute 10-year risk of dementia, by age, sex and apolipoprotein E (APOE) genotype. METHODS We obtained data from the Copenhagen General Population Study (from 2003 to 2014) and the Copenhagen City Heart Study (from 1991 to 1994 and 2001 to 2003). Participants underwent a questionnaire, physical examination and blood sampling at baseline. Diagnoses of dementia and cerebrovascular disease were obtained from the Danish National Patient Registry up to Nov. 10, 2014. RESULTS Among 104 537 individuals, the absolute 10-year risk of Alzheimer disease in 3017 women and men who were carriers of the APOE ɛ44 genotype was, respectively, 7% and 6% at age 60-69 years, 16% and 12% at age 70-79 years, and 24% and 19% at age 80 years and older. Corresponding values for all dementia were 10% and 8%, 22% and 19%, and 38% and 33%, respectively. Adjusted hazard ratios (HRs) for all dementia increased by genotype, from genotype ɛ22 to ɛ32 to ɛ33 to ɛ42 to ɛ43 to ɛ44 (p for trend < 0.001). Compared with ɛ33 carriers, ɛ44 carriers were more likely to develop Alzheimer disease (adjusted HR 8.74, 95% confidence interval [CI] 7.08-10.79), vascular dementia (adjusted HR 2.87, 95% CI 1.54-5.33), unspecified dementia (adjusted HR 4.68, 95% CI 3.74-5.85) and all dementia (adjusted HR 5.77, 95% CI 4.89-6.81). INTERPRETATION Age, sex and APOE genotype robustly identify high-risk groups for Alzheimer disease and all dementia. These groups can potentially be targeted for preventive interventions.
Collapse
Affiliation(s)
- Katrine L Rasmussen
- Department of Clinical Biochemistry (Rasmussen, Tybjærg-Hansen, Frikke-Schmidt), Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Biochemistry (Rasmussen, Nordestgaard), Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Anne Tybjærg-Hansen
- Department of Clinical Biochemistry (Rasmussen, Tybjærg-Hansen, Frikke-Schmidt), Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Biochemistry (Rasmussen, Nordestgaard), Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry (Rasmussen, Tybjærg-Hansen, Frikke-Schmidt), Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Biochemistry (Rasmussen, Nordestgaard), Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry (Rasmussen, Tybjærg-Hansen, Frikke-Schmidt), Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Biochemistry (Rasmussen, Nordestgaard), Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| |
Collapse
|
25
|
Kummer BR, Diaz I, Wu X, Aaroe AE, Chen ML, Iadecola C, Kamel H, Navi BB. Associations between cerebrovascular risk factors and parkinson disease. Ann Neurol 2019; 86:572-581. [PMID: 31464350 DOI: 10.1002/ana.25564] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 07/22/2019] [Accepted: 07/28/2019] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To determine whether cerebrovascular risk factors are associated with subsequent diagnoses of Parkinson disease, and whether these associations are similar in magnitude to those with subsequent diagnoses of Alzheimer disease. METHODS This was a retrospective cohort study using claims data from a 5% random sample of Medicare beneficiaries from 2008 to 2015. The exposures were stroke, atrial fibrillation, coronary disease, hyperlipidemia, hypertension, sleep apnea, diabetes mellitus, heart failure, peripheral vascular disease, chronic kidney disease, chronic obstructive pulmonary disease, valvular heart disease, tobacco use, and alcohol abuse. The primary outcome was a new diagnosis of idiopathic Parkinson disease. The secondary outcome was a new diagnosis of Alzheimer disease. Marginal structural Cox models adjusting for time-dependent confounding were used to characterize the association between exposures and outcomes. We also evaluated the association between cerebrovascular risk factors and subsequent renal colic (negative control). RESULTS Among 1,035,536 Medicare beneficiaries followed for a mean of 5.2 years, 15,531 (1.5%) participants were diagnosed with Parkinson disease and 81,974 (7.9%) were diagnosed with Alzheimer disease. Most evaluated cerebrovascular risk factors, including prior stroke (hazard ratio = 1.55; 95% confidence interval = 1.39-1.72), were associated with the subsequent diagnosis of Parkinson disease. The magnitudes of these associations were similar, but attenuated, to the associations between cerebrovascular risk factors and Alzheimer disease. Confirming the validity of our analytical model, most cerebrovascular risk factors were not associated with the subsequent diagnosis of renal colic. INTERPRETATION Cerebrovascular risk factors are associated with Parkinson disease, an effect comparable to their association with Alzheimer disease. ANN NEUROL 2019;86:572-581.
Collapse
Affiliation(s)
- Benjamin R Kummer
- Clinical and Translational Neuroscience Unit, Feil Family Brain and Mind Research Institute, and Department of Neurology, Weill Cornell Medicine, New York, NY
| | - Iván Diaz
- Division of Biostatistics and Epidemiology, Department of Healthcare Policy and Research, Weill Cornell Medicine, New York, NY
| | - Xian Wu
- Division of Biostatistics and Epidemiology, Department of Healthcare Policy and Research, Weill Cornell Medicine, New York, NY
| | - Ashley E Aaroe
- Clinical and Translational Neuroscience Unit, Feil Family Brain and Mind Research Institute, and Department of Neurology, Weill Cornell Medicine, New York, NY
| | - Monica L Chen
- Clinical and Translational Neuroscience Unit, Feil Family Brain and Mind Research Institute, and Department of Neurology, Weill Cornell Medicine, New York, NY
| | - Costantino Iadecola
- Clinical and Translational Neuroscience Unit, Feil Family Brain and Mind Research Institute, and Department of Neurology, Weill Cornell Medicine, New York, NY
| | - Hooman Kamel
- Clinical and Translational Neuroscience Unit, Feil Family Brain and Mind Research Institute, and Department of Neurology, Weill Cornell Medicine, New York, NY
| | - Babak B Navi
- Clinical and Translational Neuroscience Unit, Feil Family Brain and Mind Research Institute, and Department of Neurology, Weill Cornell Medicine, New York, NY
| |
Collapse
|
26
|
de Oliveira FF, de Almeida SS, Chen ES, Smith MC, Naffah-Mazzacoratti MDG, Bertolucci PHF. Lifetime Risk Factors for Functional and Cognitive Outcomes in Patients with Alzheimer's Disease. J Alzheimers Dis 2019; 65:1283-1299. [PMID: 30149448 DOI: 10.3233/jad-180303] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Lifetime risk factors for cognitive and functional decline in Alzheimer's disease (AD) are not fully understood, and were prospectively evaluated in patients with low mean schooling from São Paulo, Brazil. Consecutive outpatients with late-onset AD were assessed for APOE haplotypes and the following potential baseline predictors: gender, schooling, age at dementia onset, lifetime urban living and sanitary conditions, occupational complexity, cognitive and physical activities, cerebrovascular risk factors (obesity, lifetime alcohol use and smoking, length of arterial hypertension, diabetes mellitus, and a dyslipidemic profile), use of a pacemaker, creatinine clearance, body mass index, waist circumference, head traumas with unconsciousness, treated systemic bacterial infections, amount of surgical procedures under general anesthesia, and family history of AD. Participants were followed from October 2010 to May 2017 for baseline risk factor associations with time since dementia onset for Clinical Dementia Rating and Mini-Mental State Examination score changes. For 227 patients (154 women, 119 APOE ε 4 carriers), later AD onset (mean 73.60±6.4 years-old, earlier for APOE ε 4/ε 4 carriers, p < 0.001) was the only variable hastening all endpoints, baseline creatinine clearance and lifetime alcohol use were hazardous for earlier cognitive and functional endpoints, women had earlier cognitive endpoints only, and schooling had a cumulative protective effect over later cognitive endpoints, particularly for carriers of APOE ε 4. Exclusively for carriers of APOE ε 4, head traumas with unconsciousness were hazardous for earlier cognitive endpoints, while lifetime sanitary conditions were protective regarding later cognitive endpoints. Functional and cognitive outcomes in AD represent probable interactions between effects of brain reserve and cerebral perfusion over neurodegeneration.
Collapse
Affiliation(s)
- Fabricio Ferreira de Oliveira
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Sandro Soares de Almeida
- Department of Biophysics, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Elizabeth Suchi Chen
- Department of Morphology and Genetics, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Marilia Cardoso Smith
- Department of Morphology and Genetics, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | | | | |
Collapse
|
27
|
Korologou-Linden R, O'Keeffe L, Howe LD, Davey-Smith G, Jones HJ, Anderson EL, Stergiakouli E. Polygenic risk score for Alzheimer's disease and trajectories of cardiometabolic risk factors in children. Wellcome Open Res 2019; 4:125. [PMID: 31984241 PMCID: PMC6971849 DOI: 10.12688/wellcomeopenres.15359.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2019] [Indexed: 12/18/2022] Open
Abstract
Introduction: Cardiometabolic factors are implicated in the aetiology of Alzheimer's disease and may lie on the pathways linking genetic variants to Alzheimer's disease across the life course. We examined whether polygenic risk scores (PRS) were associated with cardiometabolic health indicators through childhood and adolescence. Methods: In 7,977 participants from the Avon Longitudinal Study of Parents and Children, we tested whether a PRS for Alzheimer's disease was associated with trajectories of cardiometabolic risk factors. We examined trajectories for height at 1-18 years; lean and fat mass at 9-18 years; systolic and diastolic blood pressure at 7-18 years; glucose and C-reactive protein at 9-18 years; insulin at 10-18 years; and high and low-density lipoproteins and triglycerides birth at 18 years. We also examined birthweight and interleukin-6 (IL-6) at age 9 years and physical activity at ages 11, 12, and 15 years. Results: No consistent associations were observed between the PRS excluding genetic variants in the apolipoprotein E gene region and cardiometabolic factors trajectories across childhood and adolescence. Conclusions: We did not detect evidence to suggest that the PRS for Alzheimer's disease acts through childhood and adolescent cardiometabolic risk factors. Further studies should examine whether these associations emerge later in adulthood when variation in cardiometabolic risk factors is likely to be greater.
Collapse
Affiliation(s)
- Roxanna Korologou-Linden
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Linda O'Keeffe
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Laura D. Howe
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - George Davey-Smith
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Hannah J. Jones
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Centre for Academic Mental Health, Population Health Sciences, University of Bristol, Bristol, UK
| | - Emma L. Anderson
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Evie Stergiakouli
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- School of Oral and Dental Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
28
|
Colín-Castelán D, Zaina S. Associations between atherosclerosis and neurological diseases, beyond ischemia-induced cerebral damage. Rev Endocr Metab Disord 2019; 20:15-25. [PMID: 30891682 DOI: 10.1007/s11154-019-09486-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neurodegeneration is traditionally viewed as a consequence of peptide accumulation in the brain, stroke and/or cerebral ischemia. Nonetheless, a number of scattered observations suggest that neurological disease and atherosclerosis may be linked by more complex mechanisms. Understanding the intricate link between atherosclerosis and neurological conditions may have a significant impact on the quality of life of the growing ageing population and of high cardiovascular risk groups in general. Epidemiological data support the notion that neurological dysfunction and atherosclerosis coexist long before any evident clinical complications of cardiovascular disease appear and may be causally linked. Baffling, often overlooked, molecular data suggest that nervous tissue-specific gene expression is relaxed specifically in the atheromatous vascular wall, and/or that a systemic dysregulation of genes involved in nervous system biology dictates a concomitant progression of neurological disease and atherosclerosis. Further epidemiological and experimental work is needed to clarify the details and clinical relevance of those complex links.
Collapse
Affiliation(s)
- Dannia Colín-Castelán
- Department of Medical Sciences, Division of Health Sciences, Campus León, University of Guanajuato, León, Guanajuato, Mexico.
| | - Silvio Zaina
- Department of Medical Sciences, Division of Health Sciences, Campus León, University of Guanajuato, León, Guanajuato, Mexico
| |
Collapse
|
29
|
Singh I, Edwards C, Duric D, Rasuly A, Musa SO, Anwar A. Dementia in an Acute Hospital Setting: Health Service Research to Profile Patient Characteristics and Predictors of Adverse Clinical Outcomes. Geriatrics (Basel) 2019; 4:geriatrics4010007. [PMID: 31023975 PMCID: PMC6473705 DOI: 10.3390/geriatrics4010007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 12/27/2018] [Accepted: 12/28/2018] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION Patients with dementia often have other associated medical co-morbidities resulting in adverse outcomes. The National Audit of Dementia (NAD) in the UK showed a wide variation in the quality and clinical care for acute dementia patients. This study aims to record the clinical profile and benchmark clinical outcomes of acute dementia patients admitted within Aneurin Bevan University Health Board, Wales (UK). METHODS This was a retrospective observational study based on analysis of the existing data for all acute dementia patients. Ethical approval was not required for this service evaluation. RESULTS In 2016, a total of 1770 dementia patients had 2474 acute admissions. We studied 1167 acute admissions (953 dementia patients) from 1st January 2016⁻30th June 2016. The mean age was 84.5 ± 7.8 years (females = 63.5%). Mean Charlson comorbidity index and the number of drugs were 6.0 ± 1.5 and 5.1 ± 2.1. 15.4% (147/953) patients were on antipsychotics. Overall mean hospital stay was 19.4 ± 27.2 days. 30-days readmission rate was 17.2% (138/800) with a mean hospital stay of 14.6 ± 17.9 days. 3.4% (32/953) patients were excluded due to a coding error. 70.3% (n = 670/953) were previously living in their own homes and only 26.3% (n = 251/953) were admitted from care homes. 59.5% patients (n = 399/670) were discharged back to their homes and 21.6% (145/670) were discharged to a new care home, which represents an approximately 1.68 times higher rate of new care home occupancy than the patients being originally admitted from a care home. Overall inpatient was 16.0% (153/953). 30-days and one-year mortality were 22.3% (213/953) and 49.2% (469/953) respectively. The observed mortality rates between patients admitted from home or from a care home were highly significant for one-year mortality (p < 0.001). The inpatient falls rate was significantly higher (1.8 times) as compared to overall general medical inpatient falls rate. CONCLUSION Acute patients with dementia have a higher risk of adverse outcomes and the impact of hospitalisation. Prompt comprehensive geriatric assessment and quality improvement initiatives are needed to improve clinical outcomes and to enhance the quality of care.
Collapse
Affiliation(s)
- Inderpal Singh
- Consultant Geriatrician, Department of Geriatric Medicine, Ysbyty Ystrad Fawr, Aneurin Bevan University Health Board, Wales CF82 7EP, UK.
| | - Chris Edwards
- Department of Dermatology, St Wollas Hospital, Aneurin Bevan University Health Board, Newport NP20 4SZ, UK.
| | - Daniel Duric
- Speciality Doctor, Department of Geriatric Medicine, Ysbyty Ystrad Fawr, Aneurin Bevan University Health Board, Wales CF82 7EP, UK.
| | - Aman Rasuly
- Speciality Doctor, Department of Geriatric Medicine, Ysbyty Ystrad Fawr, Aneurin Bevan University Health Board, Wales CF82 7EP, UK.
| | - Sabdat Oziohu Musa
- Speciality Doctor, Department of Geriatric Medicine, Ysbyty Ystrad Fawr, Aneurin Bevan University Health Board, Wales CF82 7EP, UK.
| | - Anser Anwar
- Speciality Doctor, Department of Geriatric Medicine, Ysbyty Ystrad Fawr, Aneurin Bevan University Health Board, Wales CF82 7EP, UK.
| |
Collapse
|
30
|
Pizzol FLFD, Vieira LF, Bierhals CCBK, Azzolin KDO, Paskulin LMG, Low G, Rosa NGD, Lucena ADF. Relationship between elderly stroke patient caregivers scale and nursing diagnoses. Rev Bras Enferm 2019; 72:251-258. [DOI: 10.1590/0034-7167-2018-0787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 10/13/2018] [Indexed: 11/22/2022] Open
Abstract
ABSTRACT Objective: To describe relationships between the ECPICID-AVC scale factors and the NANDA-I domains, classes, and Nursing Diagnoses (NDs). Method: Cross-mapping study between the NANDA-I taxonomy and ECPICID-AVC scale was constructed based on the eight ECPICID-AVC scale factors and the 13 NANDA-I domains. A descriptive analysis was performed to present the mapped elements. Results: Areas of similarity and intersection were found between the eight ECPICID-AVC factors and nine NANDA-I domains, 19 classes, and 72 NDs. All scale factors were mapped with the Domain 1/Health Promotion, Class 2/Health Management and the ND “Frail elderly syndrome”. Final considerations: The ECPICID-AVC scale factors were mapped with nine domains, their classes and diagnoses. This study demonstrates the importance of identifying nursing diagnoses and their relationship with factors that evaluate caregiving capacity. The ECPICID-AVC can help nurses generate nursing diagnoses regarding the caregiver’s needs and their capacities related to care to focus such needs.
Collapse
|
31
|
Zhu B, Li LX, Zhang L, Yang S, Tian Y, Guo SS, Zhang W, Zhao ZG. Correlation of PICALM polymorphism rs3851179 with Alzheimer's disease among Caucasian and Chinese populations: a meta-analysis and systematic review. Metab Brain Dis 2018; 33:1849-1857. [PMID: 30039188 DOI: 10.1007/s11011-018-0291-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 07/11/2018] [Indexed: 11/28/2022]
Abstract
The rs3851179 which located at upstream of PICALM was reported to be associated with Alzheimer's disease (AD); however, the relationship is still undefined. To gain a more precise understanding of the association, we conducted a meta-analysis: a comprehensive survey of 16 case-control studies that evaluated the role of rs3851179 gene variants in AD patients. The overall analysis revealed a significant association between the polymorphism and AD in the allelic, homozygote, heterozygote, dominant, and recessive models (p < 0.05). When stratified by ethnicity, a significant association was observed between AD development in Caucasian populations and the five-genetic models; Asian populations, however, featured a significant association in only the allelic, homozygote, and recessive models. We did not observe any influence of APOE ε4 carrier status on the incidence of AD and rs3851179 (p > 0.05). Our meta-analysis thus suggested that the PICALM rs3851179 polymorphism was associated with AD; the APOE ε4 status did not influence the relationship. Nevertheless, considering the limitations of our meta-analysis, further large-scale studies should be conducted to gain a more comprehensive understanding.
Collapse
Affiliation(s)
- Bin Zhu
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China
| | - Li-Xia Li
- Department of Geriatrics, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China
| | - Lei Zhang
- Department of Pharmacy, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100050, China
| | - Shu Yang
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China
| | - Yue Tian
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China
| | - Shan-Shan Guo
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China
| | - Wei Zhang
- Department of Geriatrics, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China.
| | - Zhi-Gang Zhao
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
32
|
Viña J, Sanz-Ros J. Alzheimer's disease: Only prevention makes sense. Eur J Clin Invest 2018; 48:e13005. [PMID: 30028503 DOI: 10.1111/eci.13005] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 07/18/2018] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease therapeutics is one of the most important endeavours in today's clinical investigation. Over more than 30 years of research, no disease-modifying treatment has been approved by either the FDA or the EMA to treat Alzheimer's disease. Recently, the evidence of pathological alterations in the brain tissue has been gathered showing that the signs of brain damage appear more than 20 years before the onset of Alzheimer's dementia. The major aim of this review is to underpin the idea that in Alzheimer's therapeutics, only prevention makes sense. It is difficult to visualise that would-be patients may be treated with endovenous administration of antibodies for several years to delay the onset of dementia. Rather, changes in lifestyle that should be specific, stratified and personalised are a likely alternative to delay the transition from asymptomatic Alzheimer's to minimal cognitive impairment and from there to dementia. These efforts are of the utmost importance. If we could delay the onset of full-blown dementia by 5 years, the number of demented patients would be almost halved. Thus, emphasis on preventive measures that can be implemented for decades must be supported. This approach, where even mild changes in cognition are of the greatest importance, cannot be underestimated in terms of both the individual and society's viewpoints.
Collapse
Affiliation(s)
- José Viña
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain.,INCLIVA Health Research Institute, Valencia, Spain.,Center for Biomedical Network Research on Frailty and Healthy Aging (CIBERFES), CIBER-ISCIII, Madrid, Spain
| | - Jorge Sanz-Ros
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain.,INCLIVA Health Research Institute, Valencia, Spain.,Center for Biomedical Network Research on Frailty and Healthy Aging (CIBERFES), CIBER-ISCIII, Madrid, Spain
| |
Collapse
|
33
|
The genetic risk of Alzheimer's disease beyond APOE ε4: systematic review of Alzheimer's genetic risk scores. Transl Psychiatry 2018; 8:166. [PMID: 30143603 PMCID: PMC6109140 DOI: 10.1038/s41398-018-0221-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 07/16/2018] [Indexed: 01/19/2023] Open
Abstract
The ε4 allele of Apolipoprotein E (APOE) is the strongest known genetic risk factor of Alzheimer's disease (AD) but does not account for the entirety of genetic risk. Genetic risk scores (GRSs) incorporating additional genetic variants have been developed to determine the genetic risk for AD, yet there is no systematic review assessing the contribution of GRSs for AD beyond the effect of APOE ε4. The purpose of this systematic PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-analyses)-based review was to summarize original research studies that have developed and validated a GRS for AD utilizing associated single nucleotide polymorphisms (SNPs). The PubMed and Web of Science databases were searched on April 6, 2018 and screening was completed on 2018 citations by two independent reviewers. Eighteen studies published between 2010 and 2018 were included in the review. All GRSs expressed significant associations or discrimination capability of AD when compared to clinically normal controls; however, GRS prediction of MCI to AD conversion was mixed. APOE ε4 status was more predictive of AD than the GRSs, although the GRSs did add to AD prediction accuracy beyond APOE ε4. GRSs might contribute to identifying genetic risk of AD beyond APOE. However, additional studies are warranted to assess the performance of GRSs in independent longitudinal cohorts.
Collapse
|
34
|
Gong S, Su BB, Tovar H, Mao C, Gonzalez V, Liu Y, Lu Y, Wang KS, Xu C. Polymorphisms Within RYR3 Gene Are Associated With Risk and Age at Onset of Hypertension, Diabetes, and Alzheimer's Disease. Am J Hypertens 2018; 31:818-826. [PMID: 29590321 DOI: 10.1093/ajh/hpy046] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 03/23/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Hypertension affects 33% of Americans while type 2 diabetes and Alzheimer's disease (AD) affect 10% of Americans, respectively. Ryanodine receptor 3 gene (RYR3) codes for the RYR which functions to release stored endoplasmic reticulum calcium ions (Ca2+) to increase intracellular Ca2+ concentration. Increasing studies demonstrate that altered levels of intracellular Ca2+ affect cardiac contraction, insulin secretion, and neurodegeneration. In this study, we investigated associations of the RYR3 genetic variants with hypertension, AD, and diabetes. METHODS Family data sets were used to explore association of RYR3 polymorphisms with risk and age at onset (AAO) of hypertension, diabetes, and AD. RESULTS Family-based association tests using generalized estimating equations (FBAT-GEE) showed several unique or shared disease-1 associated variants in the RYR3 gene. Three single nuclear polymorphisms (SNPs; rs2033610, rs2596164, and rs2278317) are significantly associated with risk for hypertension, diabetes, and AD. Two SNPs (rs4780174 and rs7498093) are significantly associated with AAO of the 3 diseases. CONCLUSIONS RYR3 variants are associated with hypertension, diabetes, and AD. Replication of these results of this gene in these 3 complex traits may help to better understand the genetic basis of calcium-signaling gene, RYR3 in association with risk and AAO of these diseases.
Collapse
Affiliation(s)
- Shaoqing Gong
- School of Public Policy and Administration, Institute of Health Policy and Administration, Xi’an Jiaotong University, Xi’an, China
| | - Brenda Bin Su
- Department of Biomedical Science, College of Medicine, University of Texas Rio Grande Valley; Chinese Medical Center, Dubai, United Arab Emirates
| | - Hugo Tovar
- Department of Health and Biomedical Sciences, College of Health Affairs, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - ChunXiang Mao
- Department of Health and Biomedical Sciences, College of Health Affairs, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - Valeria Gonzalez
- Department of Health and Biomedical Sciences, College of Health Affairs, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - Ying Liu
- Department of Biostatistics and Epidemiology, College of Public Health, East Tennessee State University, Johnson City, Tennessee, USA
| | - Yongke Lu
- Department of Health Sciences, College of Public Health, East Tennessee State University, Johnson, City, Tennessee, USA
| | - Ke-Sheng Wang
- Department of Biostatistics and Epidemiology, College of Public Health, East Tennessee State University, Johnson City, Tennessee, USA
| | - Chun Xu
- Department of Health and Biomedical Sciences, College of Health Affairs, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| |
Collapse
|
35
|
Dong S, Maniar S, Manole MD, Sun D. Cerebral Hypoperfusion and Other Shared Brain Pathologies in Ischemic Stroke and Alzheimer's Disease. Transl Stroke Res 2018; 9:238-250. [PMID: 28971348 PMCID: PMC9732865 DOI: 10.1007/s12975-017-0570-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 09/05/2017] [Accepted: 09/13/2017] [Indexed: 12/13/2022]
Abstract
Newly emerged evidence reveals that ischemic stroke and Alzheimer's disease (AD) share pathophysiological changes in brain tissue including hypoperfusion, oxidative stress, immune exhaustion, and inflammation. A mechanistic link between hypoperfusion and amyloid β accumulation can lead to cell damage as well as to motor and cognitive deficits. This review will discuss decreased cerebral perfusion and other related pathophysiological changes common to both ischemic stroke and AD, such as vascular damages, cerebral blood flow alteration, abnormal expression of amyloid β and tau proteins, as well as behavioral and cognitive deficits. Furthermore, this review highlights current treatment options and potential therapeutic targets that warrant further investigation.
Collapse
Affiliation(s)
- Shuying Dong
- Department of Pharmacology, Bengbu Medical College, Bengbu, Anhui, China
- Department of Neurology, University of Pittsburgh, S-598 South Biomedical Science Tower, 3500 Terrace St., Pittsburgh, PA, 15213, USA
| | - Shelly Maniar
- Department of Neurology, University of Pittsburgh, S-598 South Biomedical Science Tower, 3500 Terrace St., Pittsburgh, PA, 15213, USA
- Lake Erie College of Osteopathic Medicine at Seton Hill, Greensburg, Pennsylvania, 15601, USA
| | - Mioara D Manole
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, S-598 South Biomedical Science Tower, 3500 Terrace St., Pittsburgh, PA, 15213, USA.
- Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Educational and Clinical Center, Pittsburgh, PA, USA.
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW Alzheimer's disease and cerebrovascular disease (CVD) commonly co-occur. Whether CVD promotes the progression of Alzheimer's disease pathology remains a source of great interest. Recent technological developments have enabled us to examine their inter-relationship using quantifiable, biomarker-based approaches. We provide an overview of advances in understanding the relationship between vascular and Alzheimer's disease pathologies, with particular emphasis on β-amyloid and tau as measured by positron emission tomography and cerebrospinal fluid (CSF) concentration, and magnetic resonance imaging markers of small vessel disease (SVD). RECENT FINDINGS The relationship between cerebral β-amyloid and various markers of SVD has been widely studied, albeit with somewhat mixed results. Significant associations have been elucidated, particularly between β-amyloid burden and white matter hyperintensities (WMH), as well as lobar cerebral microbleeds (CMB), with additive effects on cognition. There is preliminary evidence for an association between SVD and tau burden in vivo, although compared with β-amyloid, fewer studies have examined this relationship. SUMMARY The overlap between Alzheimer's disease and cerebrovascular pathologies is now being increasingly supported by imaging and CSF biomarkers, indicating a synergistic effect of these co-pathologies on cognition. The association of WMH and CMB with Alzheimer's disease pathology does not establish direction of causality, for which long-term longitudinal studies are needed.
Collapse
|
37
|
Unschuld PG. Novel Translational Research Methodology and the Prospect to a Better Understanding of Neurodegenerative Disease. NEURODEGENER DIS 2018; 18:1-4. [PMID: 29339665 DOI: 10.1159/000486565] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- Paul G Unschuld
- Hospital for Psychogeriatric Medicine, Psychiatric University Hospital Zurich, Zurich, Switzerland.,Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| |
Collapse
|
38
|
Bennett DA, Buchman AS, Boyle PA, Barnes LL, Wilson RS, Schneider JA. Religious Orders Study and Rush Memory and Aging Project. J Alzheimers Dis 2018; 64:S161-S189. [PMID: 29865057 PMCID: PMC6380522 DOI: 10.3233/jad-179939] [Citation(s) in RCA: 705] [Impact Index Per Article: 117.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The Religious Orders Study and Rush Memory and Aging Project are both ongoing longitudinal clinical-pathologic cohort studies of aging and Alzheimer's disease (AD). OBJECTIVES To summarize progress over the past five years and its implications for understanding neurodegenerative diseases. METHODS Participants in both studies are older adults who enroll without dementia and agree to detailed longitudinal clinical evaluations and organ donation. The last review summarized findings through the end of 2011. Here we summarize progress and study findings over the past five years and discuss new directions for how these studies can inform on aging and AD in the future. RESULTS We summarize 1) findings on the relation of neurobiology to clinical AD; 2) neurobiologic pathways linking risk factors to clinical AD; 3) non-cognitive AD phenotypes including motor function and decision making; 4) the development of a novel drug discovery platform. CONCLUSION Complexity at multiple levels needs to be understood and overcome to develop effective treatments and preventions for cognitive decline and AD dementia.
Collapse
Affiliation(s)
- David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL., USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL., USA
| | - Aron S. Buchman
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL., USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL., USA
| | - Patricia A. Boyle
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL., USA
- Department of Behavioral Sciences, Rush University Medical Center, Chicago, IL., USA
| | - Lisa L. Barnes
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL., USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL., USA
- Department of Behavioral Sciences, Rush University Medical Center, Chicago, IL., USA
| | - Robert S. Wilson
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL., USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL., USA
- Department of Behavioral Sciences, Rush University Medical Center, Chicago, IL., USA
| | - Julie A Schneider
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL., USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL., USA
- Department of Pathology (Neuropathology), Rush University Medical Center, Chicago, IL., USA
| |
Collapse
|
39
|
Ibanez L, Dube U, Saef B, Budde J, Black K, Medvedeva A, Del-Aguila JL, Davis AA, Perlmutter JS, Harari O, Benitez BA, Cruchaga C. Parkinson disease polygenic risk score is associated with Parkinson disease status and age at onset but not with alpha-synuclein cerebrospinal fluid levels. BMC Neurol 2017; 17:198. [PMID: 29141588 PMCID: PMC5688622 DOI: 10.1186/s12883-017-0978-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 11/05/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The genetic architecture of Parkinson's Disease (PD) is complex and not completely understood. Multiple genetic studies to date have identified multiple causal genes and risk loci. Nevertheless, most of the expected genetic heritability remains unexplained. Polygenic risk scores (PRS) may provide greater statistical power and inform about the genetic architecture of multiple phenotypes. The aim of this study was to test the association between PRS and PD risk, age at onset and cerebrospinal fluid (CSF) biomarkers (α-synuclein, Aβ1-42, t-tau and p-tau). METHODS The weighted PRS was created using the genome-wide loci from Nalls et al., 2014 PD GWAs meta-analysis. The PRS was tested for association with PD status, age at onset and CSF biomarker levels in 829 cases and 432 controls of European ancestry. RESULTS The PRS was associated with PD status (p = 5.83×10-08) and age at onset (p = 5.70×10-07). The CSF t-tau levels showed a nominal association with the PRS (p = 0.02). However, CSF α-synuclein, amyloid beta and phosphorylated tau were not found to be associated with the PRS. CONCLUSION Our study suggests that there is an overlap in the genetic architecture of PD risk and onset, although the different loci present different weights for those phenotypes. In our dataset we found a marginal association of the PRS with CSF t-tau but not with α-synuclein CSF levels, suggesting that the genetic architecture for the CSF biomarker levels is different from that of PD risk.
Collapse
Affiliation(s)
- Laura Ibanez
- Department of Psychiatry, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA.,Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Umber Dube
- Department of Psychiatry, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA.,Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University in Saint Louis, Saint Louis, MO, USA.,Medical Scientist Training Program, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Benjamin Saef
- Department of Psychiatry, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA.,Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University in Saint Louis, Saint Louis, MO, USA
| | - John Budde
- Department of Psychiatry, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA.,Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Kathleen Black
- Department of Psychiatry, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA.,Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Alexandra Medvedeva
- Department of Psychiatry, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA.,Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Jorge L Del-Aguila
- Department of Psychiatry, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA.,Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Albert A Davis
- Department of Neurology, School of Medicine, Washington University in St Louis, Saint Louis, MO, USA
| | - Joel S Perlmutter
- Department of Neurology, School of Medicine, Washington University in St Louis, Saint Louis, MO, USA
| | - Oscar Harari
- Department of Psychiatry, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA.,Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Bruno A Benitez
- Department of Medicine, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, School of Medicine, Washington University in Saint Louis, Saint Louis, MO, USA. .,Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University in Saint Louis, Saint Louis, MO, USA.
| |
Collapse
|
40
|
Park L, Uekawa K, Garcia-Bonilla L, Koizumi K, Murphy M, Pistik R, Younkin L, Younkin S, Zhou P, Carlson G, Anrather J, Iadecola C. Brain Perivascular Macrophages Initiate the Neurovascular Dysfunction of Alzheimer Aβ Peptides. Circ Res 2017; 121:258-269. [PMID: 28515043 DOI: 10.1161/circresaha.117.311054] [Citation(s) in RCA: 164] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
RATIONALE Increasing evidence indicates that alterations of the cerebral microcirculation may play a role in Alzheimer disease, the leading cause of late-life dementia. The amyloid-β peptide (Aβ), a key pathogenic factor in Alzheimer disease, induces profound alterations in neurovascular regulation through the innate immunity receptor CD36 (cluster of differentiation 36), which, in turn, activates a Nox2-containing NADPH oxidase, leading to cerebrovascular oxidative stress. Brain perivascular macrophages (PVM) located in the perivascular space, a major site of brain Aβ collection and clearance, are juxtaposed to the wall of intracerebral resistance vessels and are a powerful source of reactive oxygen species. OBJECTIVE We tested the hypothesis that PVM are the main source of reactive oxygen species responsible for the cerebrovascular actions of Aβ and that CD36 and Nox2 in PVM are the molecular substrates of the effect. METHODS AND RESULTS Selective depletion of PVM using intracerebroventricular injection of clodronate abrogates the reactive oxygen species production and cerebrovascular dysfunction induced by Aβ applied directly to the cerebral cortex, administered intravascularly, or overproduced in the brain of transgenic mice expressing mutated forms of the amyloid precursor protein (Tg2576 mice). In addition, using bone marrow chimeras, we demonstrate that PVM are the cells expressing CD36 and Nox2 responsible for the dysfunction. Thus, deletion of CD36 or Nox2 from PVM abrogates the deleterious vascular effects of Aβ, whereas wild-type PVM reconstitute the vascular dysfunction in CD36-null mice. CONCLUSIONS The data identify PVM as a previously unrecognized effector of the damaging neurovascular actions of Aβ and unveil a new mechanism by which brain-resident innate immune cells and their receptors may contribute to the pathobiology of Alzheimer disease.
Collapse
Affiliation(s)
- Laibaik Park
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.).
| | - Ken Uekawa
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.)
| | - Lidia Garcia-Bonilla
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.)
| | - Kenzo Koizumi
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.)
| | - Michelle Murphy
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.)
| | - Rose Pistik
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.)
| | - Linda Younkin
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.)
| | - Steven Younkin
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.)
| | - Ping Zhou
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.)
| | - George Carlson
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.)
| | - Josef Anrather
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.)
| | - Costantino Iadecola
- From the Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY (L.P., K.U., L.G.B., K.K., M.M., P.Z., J.A., C.I.); McLaughlin Research Institute, Great Falls, MT (R.P., G.C.); and Mayo Clinic Jacksonville, FL (L.Y., S.Y.).
| |
Collapse
|
41
|
Kadohara K, Sato I, Kawakami K. Diabetes mellitus and risk of early-onset Alzheimer's disease: a population-based case-control study. Eur J Neurol 2017; 24:944-949. [PMID: 28503814 DOI: 10.1111/ene.13312] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 04/03/2017] [Indexed: 01/13/2023]
Abstract
BACKGROUND AND PURPOSE Previous studies have reported that diabetes is a risk factor for both all-cause and vascular dementia; however, diabetes as a risk factor for Alzheimer's disease (AD) remains controversial. Therefore, the aim was to elucidate the association between diabetes and early-onset AD. METHODS A case-control study was conducted using a population-based database that included medical and pharmacy claims and insurance eligibility data, from beneficiaries of corporate employees and their dependent family members. Cases were aged 40-64 years and were first prescribed medications for AD between 2005 and 2016. Up to four controls matched for age, sex and hospital type were included for each case. Data were analyzed using conditional logistic regression and compared between the sexes. RESULTS Data from 371 patients with AD (mean age 56.3 ± 5.3 years; 48% female) and 1484 controls were analyzed. Use of antidepressants, antipsychotics and antithrombotics during the index month was higher amongst patients with AD (19.4%, 34.5% and 11.3%, respectively) than amongst controls (2.9%, 10.3% and 7.3%, respectively). Our findings suggest no evidence for an association between diabetes and risk of early-onset AD (adjusted odds ratio 1.31; 95% confidence interval 0.90-1.92). In the subgroup analyses, adjusted odds ratios in patients with diabetes were 0.73 (95% confidence interval 0.38-1.39) and 1.68 (95% confidence interval 1.06-2.67) for female and male patients, respectively. CONCLUSIONS There is no apparent association between diabetes and risk of early-onset AD in the total study population, although a weak association was observed amongst male patients.
Collapse
Affiliation(s)
- K Kadohara
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Kyoto, Japan
| | - I Sato
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Kyoto, Japan.,The Keihanshin Consortium for Fostering the Next Generation of Global Leaders in Research (K-CONNEX), Kyoto, Japan
| | - K Kawakami
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Kyoto, Japan
| |
Collapse
|
42
|
Hefter D, Draguhn A. APP as a Protective Factor in Acute Neuronal Insults. Front Mol Neurosci 2017; 10:22. [PMID: 28210211 PMCID: PMC5288400 DOI: 10.3389/fnmol.2017.00022] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 01/16/2017] [Indexed: 12/25/2022] Open
Abstract
Despite its key role in the molecular pathology of Alzheimer’s disease (AD), the physiological function of amyloid precursor protein (APP) is unknown. Increasing evidence, however, points towards a neuroprotective role of this membrane protein in situations of metabolic stress. A key observation is the up-regulation of APP following acute (stroke, cardiac arrest) or chronic (cerebrovascular disease) hypoxic-ischemic conditions. While this mechanism may increase the risk or severity of AD, APP by itself or its soluble extracellular fragment APPsα can promote neuronal survival. Indeed, different animal models of acute hypoxia-ischemia, traumatic brain injury (TBI) and excitotoxicity have revealed protective effects of APP or APPsα. The underlying mechanisms involve APP-mediated regulation of calcium homeostasis via NMDA receptors (NMDAR), voltage-gated calcium channels (VGCC) or internal calcium stores. In addition, APP affects the expression of survival- or apoptosis-related genes as well as neurotrophic factors. In this review, we summarize the current understanding of the neuroprotective role of APP and APPsα and possible implications for future research and new therapeutic strategies.
Collapse
Affiliation(s)
- Dimitri Hefter
- Institute of Physiology and Pathophysiology, Heidelberg UniversityHeidelberg, Germany; Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg UniversityMannheim, Germany
| | - Andreas Draguhn
- Institute of Physiology and Pathophysiology, Heidelberg University Heidelberg, Germany
| |
Collapse
|