51
|
Kumral E, Zirek O. Major neurocognitive disorder followıng isolated hippocampal ischemıc lesions. J Neurol Sci 2017; 372:496-500. [DOI: 10.1016/j.jns.2016.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 10/31/2016] [Accepted: 11/02/2016] [Indexed: 11/27/2022]
|
52
|
Resende EDPF, Costa-Silva L, Carmona KC, Machado TH, Machado JCB, Guimarães HC, Barbosa MT, Teixeira AL, de Souza LC, Caramelli P. Ischemic cerebrovascular burden evaluated by magnetic resonance imaging in an elderly Brazilian community: The Pietà study. eNeurologicalSci 2016; 5:30-34. [PMID: 29430555 PMCID: PMC5803083 DOI: 10.1016/j.ensci.2016.11.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 08/08/2016] [Accepted: 11/28/2016] [Indexed: 01/18/2023] Open
Abstract
In developing countries, cardiovascular risk factors are poorly controlled, leading to high prevalence of cerebrovascular diseases. The aim of the study was to evaluate the burden of white matter lesions in magnetic resonance through the Fazekas scale in a population aged 75 + years living in the community, and to investigate possible associations between vascular lesions, cardiovascular risk factors and cognitive status. Subjects were selected from a community-based study on brain aging conducted in Caeté (Minas Gerais state), Brazil. Overall, 177 participants (112 cognitively healthy, 36 with cognitive impairment-no dementia and 29 with dementia), being 108 women, aged 79.3 ± 3.8 years, with 3.1 ± 2.9 years of educational level, underwent a 3 Tesla magnetic resonance scanner with fluid attenuated image recovery acquisition. Severity of white matter lesions was assessed through the Fazekas scale. Severe white matter lesions were present in 31.1% of the whole sample and in 25.0% of the cognitively healthy individuals. A significant association was found between severe white matter lesions and cognitive impairment (OR = 2.20, 95% CI 1.17–6.53; p = 0.021), as well as with hypertension (OR = 1.92, 95% CI 1.03–7.39; p = 0.043). In conclusion, a high prevalence of severe white matter lesions was observed in this elderly Brazilian population sample, and white matter lesions were associated with hypertension and cognitive status. Importantly, the prevalence of white matter lesions was also high in cognitively healthy subjects. The ischemic cerebrovascular burden assessed by MRI in an elderly Brazilian community is very high. MRI white matter lesions were associated with cognitive impairment and hypertension. Even in cognitively healthy individuals the prevalence of ischemic cerebrovascular lesions was high.
Collapse
Affiliation(s)
- Elisa de Paula França Resende
- Faculdade de Medicina da Universidade Federal de Minas Gerais, Av. Prof Alfredo Balena, 190, room 246, Belo Horizonte, MG 30130-100, Brazil
| | - Luciana Costa-Silva
- Faculdade de Medicina da Universidade Federal de Minas Gerais, Av. Prof Alfredo Balena, 190, room 246, Belo Horizonte, MG 30130-100, Brazil
| | - Karoline Carvalho Carmona
- Faculdade de Medicina da Universidade Federal de Minas Gerais, Av. Prof Alfredo Balena, 190, room 246, Belo Horizonte, MG 30130-100, Brazil
| | - Thais Helena Machado
- Faculdade de Medicina da Universidade Federal de Minas Gerais, Av. Prof Alfredo Balena, 190, room 246, Belo Horizonte, MG 30130-100, Brazil
| | - João Carlos Barbosa Machado
- Faculdade de Ciências Médicas de Minas Gerais, Alameda Ezequiel Dias, 275, Belo Horizonte, MG 30130-110, Brazil
| | - Henrique Cerqueira Guimarães
- Faculdade de Medicina da Universidade Federal de Minas Gerais, Av. Prof Alfredo Balena, 190, room 246, Belo Horizonte, MG 30130-100, Brazil
| | - Maira Tonidandel Barbosa
- Faculdade de Medicina da Universidade Federal de Minas Gerais, Av. Prof Alfredo Balena, 190, room 246, Belo Horizonte, MG 30130-100, Brazil
| | - Antônio Lúcio Teixeira
- Faculdade de Medicina da Universidade Federal de Minas Gerais, Av. Prof Alfredo Balena, 190, room 246, Belo Horizonte, MG 30130-100, Brazil
| | - Leonardo Cruz de Souza
- Faculdade de Medicina da Universidade Federal de Minas Gerais, Av. Prof Alfredo Balena, 190, room 246, Belo Horizonte, MG 30130-100, Brazil
| | - Paulo Caramelli
- Faculdade de Medicina da Universidade Federal de Minas Gerais, Av. Prof Alfredo Balena, 190, room 246, Belo Horizonte, MG 30130-100, Brazil
| |
Collapse
|
53
|
Ince PG, Minett T, Forster G, Brayne C, Wharton SB. Microinfarcts in an older population-representative brain donor cohort (MRC CFAS): Prevalence, relation to dementia and mobility, and implications for the evaluation of cerebral Small Vessel Disease. Neuropathol Appl Neurobiol 2016; 43:409-418. [PMID: 27664944 PMCID: PMC5516203 DOI: 10.1111/nan.12363] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 09/22/2016] [Accepted: 09/24/2016] [Indexed: 01/14/2023]
Abstract
Introduction Microinfarcts, small ischaemic foci common in ageing brain, are associated with dementia and gait dysfunction. We determined their relationship with dementia, mobility and cerebrovascular disease in an older population‐representative brain donor cohort. These data on microinfarcts were evaluated in relation to pathological assessments of clinically significant cerebral small vessel disease (SVD). Methods Microinfarcts were assessed in the MRC Cognitive Function and Ageing Study (n = 331). Nine brain areas were staged according to the number of areas affected. Results 36% of brains showed at least 1 microinfarct. Higher cortical microinfarct stage was associated with dementia at death (OR 1.41, 95% CI 1.02; 1.96, P = 0.038), whilst cortical and subcortical microinfarct stages were associated with impaired mobility (OR 1.36, 95% CI 1.05–1.74; P 0.018) and falls (OR 1.96, 95% CI 1.11–3.43; P = 0.02). Adding data on microinfarcts to a definition of SVD, based on white matter lesions (WMLs), lacunes and significant arteriosclerosis, were assessed by comparing area under ROC curve (AUC) with and without microinfarcts. SVD was significantly related to dementia status with or without inclusion of microinfarcts. Modelling potential pathological definitions of SVD to predict dementia or impaired mobility indicated optimal prediction using combined assessment of WMLs, lacunes and microinfarcts. Conclusion Cortical (dementia) and subcortical microinfarcts (impaired mobility) are related to diverse clinical outcomes. Optimal pathological assessment of significant SVD in brain ageing is achieved based on WMLs, lacunes and microinfarcts and may not require subjective assessment of the extent and severity of arteriosclerosis.
Collapse
Affiliation(s)
- P G Ince
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - T Minett
- Institute of Public Health, University of Cambridge, Cambridge, UK.,Department of Radiology, University of Cambridge, Cambridge, UK
| | - G Forster
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - C Brayne
- Institute of Public Health, University of Cambridge, Cambridge, UK
| | - S B Wharton
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | | |
Collapse
|
54
|
Chen C, Homma A, Mok VCT, Krishnamoorthy E, Alladi S, Meguro K, Abe K, Dominguez J, Marasigan S, Kandiah N, Kim SY, Lee DY, De Silva HA, Yang YH, Pai MC, Senanarong V, Dash A. Alzheimer's disease with cerebrovascular disease: current status in the Asia-Pacific region. J Intern Med 2016; 280:359-74. [PMID: 26992016 DOI: 10.1111/joim.12495] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND There is growing awareness of the coexistence of Alzheimer's disease and cerebrovascular disease (AD+CVD), however, due to lack of well-defined criteria and treatment guidelines AD+CVD may be underdiagnosed in Asia. METHODS Sixteen dementia specialists from nine Asia Pacific countries completed a survey in September 2014 and met in November 2014 to review the epidemiology, diagnosis and treatment of AD+CVD in Asia. A consensus was reached by discussion, with evidence provided by published studies when available. RESULTS AD accounts for up to 60% and AD+CVD accounts for 10-20% of all dementia cases in Asia. The reasons for underdiagnosis of AD+CVD include lack of awareness as a result of a lack of diagnostic criteria, misdiagnosis as vascular dementia or AD, lack of diagnostic facilities, resource constraints and cost of investigations. There is variability in the tools used to diagnose AD+CVD in clinical practice. Diagnosis of AD+CVD should be performed in a stepwise manner of clinical evaluation followed by neuroimaging. Dementia patients should be assessed for cognition, behavioural and psychological symptoms, functional staging and instrumental activities of daily living. Neuroimaging should be performed using computed tomography or magnetic resonance imaging. The treatment goals are to stabilize or slow progression as well as to reduce behavioural and psychological symptoms, improve quality of life and reduce disease burden. First-line therapy is usually an acetylcholinesterase inhibitor such as donepezil. CONCLUSION AD+CVD is likely to be under-recognised in Asia. Further research is needed to establish the true prevalence of this treatable and potentially preventable disease.
Collapse
Affiliation(s)
- C Chen
- Department of Pharmacology, National University of Singapore, Singapore, Singapore. .,Memory Aging and Cognition Center, National University Health System, Singapore, Singapore.
| | - A Homma
- Research Institute for Dementia Care, Tokyo, Japan
| | - V C T Mok
- Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | | | - S Alladi
- Department of Neurology, Nizam's Institute of Medical Sciences, Hyderabad, India
| | - K Meguro
- Division of Geriatric Behavioral Neurology, CYRIC, Tohoku University, Sendai, Japan
| | - K Abe
- Department of Neurology, Okayama University, Okayama, Japan
| | - J Dominguez
- Memory Center, St Luke's Medical Center, Quezon City, Philippines
| | - S Marasigan
- Department of Neurology and Psychiatry, University of Santo Tomas Hospital, Manila, Philippines
| | - N Kandiah
- Department of Neurology, National Neuroscience Institute and Duke-NUS Singapore, Singapore, Singapore
| | - S Y Kim
- Department of Neurology, Seoul National University College of Medicine, Seoul, Korea.,Neurocognitive Behavior Center, Seoul National University Bundang Hospital, Seoul, Korea
| | - D Y Lee
- Department of Psychiatry, Seoul National University, College of Medicine, Seoul, Korea
| | - H A De Silva
- Clinical Trials Unit, Department of Pharmacology, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka.
| | - Y-H Yang
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan.,Master's Program in Neurology, Faculty of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - M-C Pai
- Division of Behavioral Neurology, Department of Neurology, Alzheimer's Disease Research Center, Medical College and Hospital, National Cheng Kung University, Tainan City, Taiwan
| | - V Senanarong
- Division of Neurology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - A Dash
- Eisai Co. Ltd, Mumbai, India
| |
Collapse
|
55
|
Justice JN, Johnson LC, DeVan AE, Cruickshank-Quinn C, Reisdorph N, Bassett CJ, Evans TD, Brooks FA, Bryan NS, Chonchol MB, Giordano T, McQueen MB, Seals DR. Improved motor and cognitive performance with sodium nitrite supplementation is related to small metabolite signatures: a pilot trial in middle-aged and older adults. Aging (Albany NY) 2016; 7:1004-21. [PMID: 26626856 PMCID: PMC4694069 DOI: 10.18632/aging.100842] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Advancing age is associated with reductions in nitric oxide bioavailability and changes in metabolic activity, which are implicated in declines in motor and cognitive function. In preclinical models, sodium nitrite supplementation (SN) increases plasma nitrite and improves motor function, whereas other nitric oxide-boosting agents improve cognitive function. This pilot study was designed to translate these findings to middle-aged and older (MA/O) humans to provide proof-of-concept support for larger trials. SN (10 weeks, 80 or 160 mg/day capsules, TheraVasc, Inc.) acutely and chronically increased plasma nitrite and improved performance on measures of motor and cognitive outcomes (all p<0.05 or better) in healthy MA/O adults (62 ± 7 years). Untargeted metabolomics analysis revealed that SN significantly altered 33 (160 mg/day) to 45 (80 mg/day) different metabolites, 13 of which were related to changes in functional outcomes; baseline concentrations of 99 different metabolites predicted functional improvements with SN. This pilot study provides the first evidence that SN improves aspects of motor and cognitive function in healthy MA/O adults, and that these improvements are associated with, and predicted by, the plasma metabolome. Our findings provide the necessary support for larger clinical trials on this promising pharmacological strategy for preserving physiological function with aging.
Collapse
Affiliation(s)
- Jamie N Justice
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Lawrence C Johnson
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Allison E DeVan
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Charmion Cruickshank-Quinn
- Integrated Department of Immunology, University of Colorado Anschutz Medical Campus and National Jewish Hospital, Denver, CO 80045, USA
| | - Nichole Reisdorph
- Integrated Department of Immunology, University of Colorado Anschutz Medical Campus and National Jewish Hospital, Denver, CO 80045, USA
| | - Candace J Bassett
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Trent D Evans
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Forrest A Brooks
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | | | - Michel B Chonchol
- Division of Renal Diseases and Hypertension, University of Colorado Denver, Aurora, CO 80045, USA
| | | | - Matthew B McQueen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
56
|
Skrobot OA, Attems J, Esiri M, Hortobágyi T, Ironside JW, Kalaria RN, King A, Lammie GA, Mann D, Neal J, Ben-Shlomo Y, Kehoe PG, Love S. Vascular cognitive impairment neuropathology guidelines (VCING): the contribution of cerebrovascular pathology to cognitive impairment. Brain 2016; 139:2957-2969. [DOI: 10.1093/brain/aww214] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 07/03/2016] [Indexed: 01/01/2023] Open
|
57
|
Yang D, Cabral D, Gaspard EN, Lipton RB, Rundek T, Derby CA. Cerebral Hemodynamics in the Elderly: A Transcranial Doppler Study in the Einstein Aging Study Cohort. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2016; 35:1907-14. [PMID: 27417737 PMCID: PMC5500193 DOI: 10.7863/ultra.15.10040] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 12/07/2015] [Indexed: 05/19/2023]
Abstract
OBJECTIVES We sought to describe the relationship between age, sex, and race/ethnicity with transcranial Doppler hemodynamic characteristics from major intracerebral arterial segments in a large elderly population with varying demographics. METHODS We analyzed 369 stroke-free participants aged 70 years and older from the Einstein Aging Study. Single-gate, nonimaging transcranial Doppler sonography, a noninvasive sonographic technique that assesses real-time cerebrovascular hemodynamics, was used to interrogate 9 cerebral arterial segments. Individual Doppler spectra and cerebral blood flow velocities were acquired, and the pulsatility index and resistive index were calculated by the device's automated waveform-tracking function. Multiple linear regression models were used to examine the independent associations of age, sex, and race/ethnicity with transcranial Doppler measures, adjusting for hypertension, history of myocardial infarction or revascularization, and history of diabetes. RESULTS Among enrolled participants, 303 individuals had at least 1 vessel insonated (mean age [SD], 80 [6] years; 63% women; 58% white; and 32% black). With age, transcranial Doppler measures of mean blood flow velocity were significantly decreased in the basilar artery (P = .001) and posterior cerebral artery (right, P = .003; left, P = .02). Pulsatility indices increased in the left middle cerebral artery (P = .01) and left anterior cerebral artery (P = .03), and the resistive index was increased in the left middle cerebral artery (P = .007) with age. Women had higher pulsatility and resistive indices compared to men in several vessels. CONCLUSIONS We report a decreased mean blood flow velocity and weakly increased arterial pulsatility and resistance with aging in a large elderly stroke-free population. These referential trends in cerebrovascular hemodynamics may carry important implications in vascular diseases associated with advanced age, increased risk of cerebrovascular disease, cognitive decline, and dementia.
Collapse
Affiliation(s)
- Dixon Yang
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, Florida USA
| | - Digna Cabral
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, Florida USA
| | - Emmanuel N Gaspard
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, New York USA
| | - Richard B Lipton
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, New York USA, Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York USA
| | - Tatjana Rundek
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, Florida USA
| | - Carol A Derby
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, New York USA, Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York USA
| |
Collapse
|
58
|
Abstract
Vascular dementia (VaD) is a major contributor to the dementia syndrome and is described as having problems with reasoning, planning, judgment, and memory caused by impaired blood flow to the brain and damage to the blood vessels resulting from events such as stroke. There are a variety of etiologies that contribute to the development of vascular cognitive impairment and VaD, and these are often associated with other dementia-related pathologies such as Alzheimer disease. The diagnosis of VaD is difficult due to the number and types of lesions and their locations in the brain. Factors that increase the risk of vascular diseases such as stroke, high blood pressure, high cholesterol, and smoking also raise the risk of VaD. Therefore, controlling these risk factors can help lower the chances of developing VaD. This update describes the subtypes of VaD, with details of their complex presentation, associated pathological lesions, and issues with diagnosis, prevention, and treatment.
Collapse
Affiliation(s)
- Ayesha Khan
- Wolfson Centre for Age Related Diseases, Guys Campus, London, United Kingdom of Great Britain and Northern Ireland Institute of NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Raj N Kalaria
- Institute for Ageing and Health, Wolfson Research Centre, Campus for Ageing & Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Anne Corbett
- Wolfson Centre for Age Related Diseases, Guys Campus, London, United Kingdom of Great Britain and Northern Ireland
| | - Clive Ballard
- Wolfson Centre for Age Related Diseases, Guys Campus, London, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
59
|
Nelson PT, Trojanowski JQ, Abner EL, Al-Janabi OM, Jicha GA, Schmitt FA, Smith CD, Fardo DW, Wang WX, Kryscio RJ, Neltner JH, Kukull WA, Cykowski MD, Van Eldik LJ, Ighodaro ET. "New Old Pathologies": AD, PART, and Cerebral Age-Related TDP-43 With Sclerosis (CARTS). J Neuropathol Exp Neurol 2016; 75:482-98. [PMID: 27209644 PMCID: PMC6366658 DOI: 10.1093/jnen/nlw033] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Indexed: 12/12/2022] Open
Abstract
The pathology-based classification of Alzheimer's disease (AD) and other neurodegenerative diseases is a work in progress that is important for both clinicians and basic scientists. Analyses of large autopsy series, biomarker studies, and genomics analyses have provided important insights about AD and shed light on previously unrecognized conditions, enabling a deeper understanding of neurodegenerative diseases in general. After demonstrating the importance of correct disease classification for AD and primary age-related tauopathy, we emphasize the public health impact of an underappreciated AD "mimic," which has been termed "hippocampal sclerosis of aging" or "hippocampal sclerosis dementia." This pathology affects >20% of individuals older than 85 years and is strongly associated with cognitive impairment. In this review, we provide an overview of current hypotheses about how genetic risk factors (GRN, TMEM106B, ABCC9, and KCNMB2), and other pathogenetic influences contribute to TDP-43 pathology and hippocampal sclerosis. Because hippocampal sclerosis of aging affects the "oldest-old" with arteriolosclerosis and TDP-43 pathologies that extend well beyond the hippocampus, more appropriate terminology for this disease is required. We recommend "cerebral age-related TDP-43 and sclerosis" (CARTS). A detailed case report is presented, which includes neuroimaging and longitudinal neurocognitive data. Finally, we suggest a neuropathology-based diagnostic rubric for CARTS.
Collapse
Affiliation(s)
- Peter T Nelson
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC).
| | - John Q Trojanowski
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Erin L Abner
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Omar M Al-Janabi
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Gregory A Jicha
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Frederick A Schmitt
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Charles D Smith
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - David W Fardo
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Wang-Xia Wang
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Richard J Kryscio
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Janna H Neltner
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Walter A Kukull
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Matthew D Cykowski
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Linda J Van Eldik
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Eseosa T Ighodaro
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| |
Collapse
|
60
|
Neuropathological diagnosis of vascular cognitive impairment and vascular dementia with implications for Alzheimer's disease. Acta Neuropathol 2016; 131:659-85. [PMID: 27062261 PMCID: PMC4835512 DOI: 10.1007/s00401-016-1571-z] [Citation(s) in RCA: 260] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/23/2016] [Accepted: 03/24/2016] [Indexed: 12/16/2022]
Abstract
Vascular dementia (VaD) is recognised as a neurocognitive disorder, which is explained by numerous vascular causes in the general absence of other pathologies. The heterogeneity of cerebrovascular disease makes it challenging to elucidate the neuropathological substrates and mechanisms of VaD as well as vascular cognitive impairment (VCI). Consensus and accurate diagnosis of VaD relies on wide-ranging clinical, neuropsychometric and neuroimaging measures with subsequent pathological confirmation. Pathological diagnosis of suspected clinical VaD requires adequate postmortem brain sampling and rigorous assessment methods to identify important substrates. Factors that define the subtypes of VaD include the nature and extent of vascular pathologies, degree of involvement of extra and intracranial vessels and the anatomical location of tissue changes. Atherosclerotic and cardioembolic diseases appear the most common substrates of vascular brain injury or infarction. Small vessel disease characterised by arteriolosclerosis and lacunar infarcts also causes cortical and subcortical microinfarcts, which appear to be the most robust substrates of cognitive impairment. Diffuse WM changes with loss of myelin and axonal abnormalities are common to almost all subtypes of VaD. Medial temporal lobe and hippocampal atrophy accompanied by variable hippocampal sclerosis are also features of VaD as they are of Alzheimer’s disease. Recent observations suggest that there is a vascular basis for neuronal atrophy in both the temporal and frontal lobes in VaD that is entirely independent of any Alzheimer pathology. Further knowledge on specific neuronal and dendro-synaptic changes in key regions resulting in executive dysfunction and other cognitive deficits, which define VCI and VaD, needs to be gathered. Hereditary arteriopathies such as cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy or CADASIL have provided insights into the mechanisms of dementia associated with cerebral small vessel disease. Greater understanding of the neurochemical and molecular investigations is needed to better define microvascular disease and vascular substrates of dementia. The investigation of relevant animal models would be valuable in exploring the pathogenesis as well as prevention of the vascular causes of cognitive impairment.
Collapse
|
61
|
Love S, Miners JS. Cerebrovascular disease in ageing and Alzheimer's disease. Acta Neuropathol 2016; 131:645-58. [PMID: 26711459 PMCID: PMC4835514 DOI: 10.1007/s00401-015-1522-0] [Citation(s) in RCA: 203] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 12/03/2015] [Accepted: 12/07/2015] [Indexed: 12/14/2022]
Abstract
Cerebrovascular disease (CVD) and Alzheimer’s disease (AD) have more in common than their association with ageing. They share risk factors and overlap neuropathologically. Most patients with AD have Aβ amyloid angiopathy and degenerative changes affecting capillaries, and many have ischaemic parenchymal abnormalities. Structural vascular disease contributes to the ischaemic abnormalities in some patients with AD. However, the stereotyped progression of hypoperfusion in this disease, affecting first the precuneus and cingulate gyrus, then the frontal and temporal cortex and lastly the occipital cortex, suggests that other factors are more important, particularly in early disease. Whilst demand for oxygen and glucose falls in late disease, functional MRI, near infrared spectroscopy to measure the saturation of haemoglobin by oxygen, and biochemical analysis of myelin proteins with differential susceptibility to reduced oxygenation have all shown that the reduction in blood flow in AD is primarily a problem of inadequate blood supply, not reduced metabolic demand. Increasing evidence points to non-structural vascular dysfunction rather than structural abnormalities of vessel walls as the main cause of cerebral hypoperfusion in AD. Several mediators are probably responsible. One that is emerging as a major contributor is the vasoconstrictor endothelin-1 (EDN1). Whilst there is clearly an additive component to the clinical and pathological effects of hypoperfusion and AD, experimental and clinical observations suggest that the disease processes also interact mechanistically at a cellular level in a manner that exacerbates both. The elucidation of some of the mechanisms responsible for hypoperfusion in AD and for the interactions between CVD and AD has led to the identification of several novel therapeutic approaches that have the potential to ameliorate ischaemic damage and slow the progression of neurodegenerative disease.
Collapse
Affiliation(s)
- Seth Love
- Institute of Clinical Neurosciences, School of Clinical Sciences, Learning and Research Level 2, Southmead Hospital, University of Bristol, Bristol, BS10 5NB, UK.
| | - J Scott Miners
- Institute of Clinical Neurosciences, School of Clinical Sciences, Learning and Research Level 2, Southmead Hospital, University of Bristol, Bristol, BS10 5NB, UK
| |
Collapse
|
62
|
2016 Alzheimer's disease facts and figures. Alzheimers Dement 2016. [DOI: 10.1016/j.jalz.2016.03.001 and (select 8682 from (select(sleep(5)))aqxj)] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
63
|
2016 Alzheimer's disease facts and figures. Alzheimers Dement 2016. [DOI: 10.1016/j.jalz.2016.03.001 order by 1-- rkdf] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
64
|
2016 Alzheimer's disease facts and figures. Alzheimers Dement 2016. [DOI: 10.1016/j.jalz.2016.03.001 waitfor delay '0:0:5'-- bmov] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
65
|
|
66
|
2016 Alzheimer's disease facts and figures. Alzheimers Dement 2016. [DOI: 10.1016/j.jalz.2016.03.001 order by 1-- bcpd] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
67
|
|
68
|
|
69
|
|
70
|
|
71
|
|
72
|
|
73
|
2016 Alzheimer's disease facts and figures. Alzheimers Dement 2016. [DOI: 10.1016/j.jalz.2016.03.001 waitfor delay '0:0:5'] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
74
|
2016 Alzheimer's disease facts and figures. Alzheimers Dement 2016. [DOI: 10.1016/j.jalz.2016.03.001 and 2364=4691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
75
|
|
76
|
2016 Alzheimer's disease facts and figures. Alzheimers Dement 2016. [DOI: 10.1016/j.jalz.2016.03.001 and 8336=8336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
77
|
2016 Alzheimer's disease facts and figures. Alzheimers Dement 2016. [DOI: 10.1016/j.jalz.2016.03.001 and 8336=8336-- yvja] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
78
|
|
79
|
2016 Alzheimer's disease facts and figures. Alzheimers Dement 2016. [DOI: 10.1016/j.jalz.2016.03.001 and 9592=(select 9592 from pg_sleep(5))-- pgrd] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
80
|
2016 Alzheimer's disease facts and figures. Alzheimers Dement 2016. [DOI: 10.1016/j.jalz.2016.03.001 and 9592=(select 9592 from pg_sleep(5))] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
81
|
2016 Alzheimer's disease facts and figures. Alzheimers Dement 2016. [DOI: 10.1016/j.jalz.2016.03.001 and (select 8682 from (select(sleep(5)))aqxj)-- zwlx] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
82
|
2016 Alzheimer's disease facts and figures. Alzheimers Dement 2016. [DOI: 10.1016/j.jalz.2016.03.001 and 5109=2486-- lenk] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
83
|
|
84
|
Zheng L, Vinters HV, Mack WJ, Weiner MW, Chui HC. Differential effects of ischemic vascular disease and Alzheimer's disease on brain atrophy and cognition. J Cereb Blood Flow Metab 2016; 36:204-15. [PMID: 26126864 PMCID: PMC4758550 DOI: 10.1038/jcbfm.2015.152] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 05/27/2015] [Accepted: 05/28/2015] [Indexed: 11/09/2022]
Abstract
We previously reported that pathologic measures of arteriosclerosis (AS), cerebral infarction, and Alzheimer’s disease (AD) are independently correlated with cortical gray matter (CGM) atrophy measured by in vivo magnetic resonance imaging (MRI). Here, we use path analyses to model the associations between these three pathology measures and cognitive impairment, as mediated by CGM atrophy, after controlling for age and education. In this sample of 116 elderly persons followed longitudinally to autopsy (ischemic vascular disease (IVD) program project), differential patterns were observed between AS and atrophy/cognition versus AD and atrophy/cognition. The total effect of AD pathology on global cognition (β = -0.61, s.e. = 0.06) was four times stronger than that of AS (β = -0.15, s.e. = 0.08). The effect of AS on cognition appears to occur through cerebral infarction and CGM atrophy (β = -0.13, s.e. = 0.04). In contrast, the effects of AD pathology on global cognition (β = -0.50, s.e. = 0.07) occur through a direct pathway that is five times stronger than the indirect pathway acting through CGM atrophy (β = -0.09, s.e. = 0.03). The strength of this direct AD pathway was not significantly mitigated by adding hippocampal volume to the model. AD pathology affects cognition not only through brain atrophy, but also via an unmeasured pathway that could be related to synaptic dysfunction before the development of cortical atrophy.
Collapse
Affiliation(s)
- Ling Zheng
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Harry V Vinters
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Wendy J Mack
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Michael W Weiner
- Departments of Medicine, Neurology, and Radiology, University of California San Francisco, San Francisco, California, USA
| | - Helena C Chui
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | | |
Collapse
|
85
|
Abstract
This report discusses the public health impact of Alzheimer’s disease (AD), including incidence and prevalence, mortality rates, costs of care and the overall effect on caregivers and society. It also examines the challenges encountered by health care providers when disclosing an AD diagnosis to patients and caregivers. An estimated 5.3 million Americans have AD; 5.1 million are age 65 years, and approximately 200,000 are age <65 years and have younger onset AD. By mid-century, the number of people living with AD in the United States is projected to grow by nearly 10 million, fueled in large part by the aging baby boom generation. Today, someone in the country develops AD every 67 seconds. By 2050, one new case of AD is expected to develop every 33 seconds, resulting in nearly 1 million new cases per year, and the estimated prevalence is expected to range from 11 million to 16 million. In 2013, official death certificates recorded 84,767 deaths from AD, making AD the sixth leading cause of death in the United States and the fifth leading cause of death in Americans age 65 years. Between 2000 and 2013, deaths resulting from heart disease, stroke and prostate cancer decreased 14%, 23% and 11%, respectively, whereas deaths from AD increased 71%. The actual number of deaths to which AD contributes (or deaths with AD) is likely much larger than the number of deaths from AD recorded on death certificates. In 2015, an estimated 700,000 Americans age 65 years will die with AD, and many of them will die from complications caused by AD. In 2014, more than 15 million family members and other unpaid caregivers provided an estimated 17.9 billion hours of care to people with AD and other dementias, a contribution valued at more than $217 billion. Average per-person Medicare payments for services to beneficiaries age 65 years with AD and other dementias are more than two and a half times as great as payments for all beneficiaries without these conditions, and Medicaid payments are 19 times as great. Total payments in 2015 for health care, long-term care and hospice services for people age 65 years with dementia are expected to be $226 billion. Among people with a diagnosis of AD or another dementia, fewer than half report having been told of the diagnosis by their health care provider. Though the benefits of a prompt, clear and accurate disclosure of an AD diagnosis are recognized by the medical profession, improvements to the disclosure process are needed. These improvements may require stronger support systems for health care providers and their patients.
Collapse
|
86
|
Sancesario GM, Bernardini S. How many biomarkers to discriminate neurodegenerative dementia? Crit Rev Clin Lab Sci 2015; 52:314-26. [PMID: 26292074 DOI: 10.3109/10408363.2015.1051658] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A number of cerebrospinal fluid (CSF) biomarkers are currently used for the diagnosis of dementia. Opposite changes in the level of amyloid-β(1-42) versus total tau and phosphorylated-tau181 in the CSF reflect the specific pathology of Alzheimer's disease (AD) in the brain. This panel of biomarkers has proven to be effective to differentiate AD from controls and from the major types of neurodegenerative dementia, and to evaluate the progression from mild cognitive impairment to AD. In the absence of specific biomarkers reflecting the pathologies of the other most common forms of dementia, such as Lewy Body disease, Frontotemporal lobar degeneration, Creutzfeldt-Jakob disease, etc., the evaluation of biomarkers of AD pathology is used, attempting to exclude rather than to confirm AD. Other biomarkers included in the common clinical practice do not clearly relate to the underlying pathology: progranulin (PGRN) is a selective marker of frontotemporal dementia with mutations in the PGRN gene; the 14-3-3 protein is a highly sensitive and specific marker for Creutzfeldt-Jakob disease, but has to be used carefully in differentiating rapid progressive dementia; and α-synuclein is an emerging candidate biomarker of the different forms of synucleinopathy. This review summarizes several biomarkers of neurodegenerative dementia validated based on the neuropathological processes occurring in brain tissue. Notwithstanding the paucity of pathologically validated biomarkers and their high analytical variability, the combinations of these biomarkers may well represent a key and more precise analytical and diagnostic tool in the complex plethora of degenerative dementia.
Collapse
Affiliation(s)
- Giulia M Sancesario
- a Department of Clinical and Behavioural Neurology , Santa Lucia Foundation, IRCCS , Rome , Italy and
| | - Sergio Bernardini
- b Department of Experimental Medicine and Surgery , Tor Vergata University of Rome , Rome , Italy
| |
Collapse
|
87
|
Wharton SB, Simpson JE, Brayne C, Ince PG. Age-associated white matter lesions: the MRC Cognitive Function and Ageing Study. Brain Pathol 2015; 25:35-43. [PMID: 25521175 DOI: 10.1111/bpa.12219] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 10/08/2014] [Indexed: 12/11/2022] Open
Abstract
Cerebral white matter lesions (WML) are common in the aging brain and are associated with dementia and depression. They are associated with vascular risk factors and small vessel disease, suggesting an ischemic origin, but recent pathology studies suggest a more complex pathogenesis. Studies using samples from the population-representative Medical Research Council Cognitive Function and Ageing Study neuropathology cohort used post-mortem magnetic resonance imaging to identify WML for further study. Expression of hypoxia-related molecules and other injury and protective cellular pathways in candidate immunohistochemical and gene expression microarray studies support a role for hypoxia/ischemia. However, these approaches also suggest that immune activation, blood-brain barrier dysfunction, altered cell metabolic pathways and glial cell injury contribute to pathogenesis. These abnormalities are not confined to WML, but are also found in apparently normal white matter in brains with lesions, suggesting a field effect of white matter abnormality within which lesions arise. WML are an active pathology with a complex pathogenesis that may potentially offer a number of primary and secondary intervention targets.
Collapse
Affiliation(s)
- Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | | | | | | |
Collapse
|
88
|
Love S, Miners JS. White matter hypoperfusion and damage in dementia: post-mortem assessment. Brain Pathol 2015; 25:99-107. [PMID: 25521180 DOI: 10.1111/bpa.12223] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 10/08/2014] [Indexed: 01/19/2023] Open
Abstract
Neuroimaging has revealed a range of white matter abnormalities that are common in dementia, some that predict cognitive decline. The abnormalities may result from structural diseases of the cerebral vasculature, such as arteriolosclerosis and amyloid angiopathy, but can also be caused by nonstructural vascular abnormalities (eg, of vascular contractility or permeability), neurovascular instability or extracranial cardiac or vascular disease. Conventional histopathological assessment of the white matter has tended to conflate morphological vascular abnormalities with changes that reflect altered interstitial fluid dynamics or white matter ischemic damage, even though the latter may be of extracranial or nonstructural etiology. However, histopathology is being supplemented by biochemical approaches, including the measurement of proteins involved in the molecular responses to brain ischemia, myelin proteins differentially susceptible to ischemic damage, vessel-associated proteins that allow rapid measurement of microvessel density, markers of blood-brain barrier dysfunction and axonal injury, and mediators of white matter damage. By combining neuroimaging with histopathology and biochemical analysis, we can provide reproducible, quantitative data on the severity of white matter damage, and information on its etiology and pathogenesis. Together these have the potential to inform and improve treatment, particularly in forms of dementia to which white matter hypoperfusion makes a significant contribution.
Collapse
Affiliation(s)
- Seth Love
- Dementia Research Group, Institute of Clinical Neurosciences, University of Bristol, Learning and Research Level 2, Southmead Hospital, Bristol, UK
| | | |
Collapse
|
89
|
Cerebral amyloid angiopathy and its co-occurrence with Alzheimer's disease and other cerebrovascular neuropathologic changes. Neurobiol Aging 2015; 36:2702-8. [PMID: 26239176 DOI: 10.1016/j.neurobiolaging.2015.06.028] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 06/29/2015] [Accepted: 06/30/2015] [Indexed: 11/21/2022]
Abstract
We examined the relationship between cerebral amyloid angiopathy (CAA), Alzheimer's disease neuropathologic changes, other vascular brain pathologies, and cognition in a large multicenter autopsy sample. Data were obtained from the National Alzheimer's Coordinating Center on autopsied subjects (N = 3976) who died between 2002 and 2012. Descriptive statistics and multivariable regression models estimated the associations between CAA and other pathologies, and between CAA severity and cognitive test scores proximal to death. CAA tended to co-occur with Alzheimer's disease neuropathologic changes but a minority of cases were discrepant. CAA was absent in 22% (n = 520) of subjects with frequent neuritic plaques but present in 20.9% (n = 91) of subjects with no neuritic plaques. In subjects with no/sparse neuritic plaques, nonhemorrhagic brain infarcts were more common in those with CAA pathology than without (p = 0.007). In subjects without the APOE ε4 allele, CAA severity was associated with lower cognition proximal to death, factoring in other pathologies. The presence of CAA in patients without Alzheimer's disease may indicate a distinct cerebrovascular condition.
Collapse
|
90
|
Holland PR, Searcy JL, Salvadores N, Scullion G, Chen G, Lawson G, Scott F, Bastin ME, Ihara M, Kalaria R, Wood ER, Smith C, Wardlaw JM, Horsburgh K. Gliovascular disruption and cognitive deficits in a mouse model with features of small vessel disease. J Cereb Blood Flow Metab 2015; 35:1005-14. [PMID: 25669904 PMCID: PMC4640247 DOI: 10.1038/jcbfm.2015.12] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 01/12/2015] [Accepted: 01/13/2015] [Indexed: 01/23/2023]
Abstract
Cerebral small vessel disease (SVD) is a major cause of age-related cognitive impairment and dementia. The pathophysiology of SVD is not well understood and is hampered by a limited range of relevant animal models. Here, we describe gliovascular alterations and cognitive deficits in a mouse model of sustained cerebral hypoperfusion with features of SVD (microinfarcts, hemorrhage, white matter disruption) induced by bilateral common carotid stenosis. Multiple features of SVD were determined on T2-weighted and diffusion-tensor magnetic resonance imaging scans and confirmed by pathologic assessment. These features, which were absent in sham controls, included multiple T2-hyperintense infarcts and T2-hypointense hemosiderin-like regions in subcortical nuclei plus increased cerebral atrophy compared with controls. Fractional anisotropy was also significantly reduced in several white matter structures including the corpus callosum. Investigation of gliovascular changes revealed a marked increase in microvessel diameter, vascular wall disruption, fibrinoid necrosis, hemorrhage, and blood-brain barrier alterations. Widespread reactive gliosis, including displacement of the astrocytic water channel, aquaporin 4, was observed. Hypoperfused mice also demonstrated deficits in spatial working and reference memory tasks. Overall, gliovascular disruption is a prominent feature of this mouse, which could provide a useful model for early-phase testing of potential SVD treatment strategies.
Collapse
Affiliation(s)
- Philip R Holland
- Centre for Neuroregeneration, Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - James L Searcy
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh, UK
| | | | - Gillian Scullion
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh, UK
| | - Guiquan Chen
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh, UK
| | - Greig Lawson
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh, UK
| | - Fiona Scott
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh, UK
| | - Mark E Bastin
- Centre for Cognitive Ageing and Cognitive Epidemiology, Scottish Imaging Network, A Platform for Scientific Collaboration (SINAPSE), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Masafumi Ihara
- Department of Stroke and Cerebrovascular Diseases, National Cerebral and Cardiovascular Center Hospital, Osaka, Japan
| | - Rajesh Kalaria
- Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
| | - Emma R Wood
- Centre for Cognitive and Neural Systems and Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Colin Smith
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Joanna M Wardlaw
- Centre for Cognitive Ageing and Cognitive Epidemiology, Scottish Imaging Network, A Platform for Scientific Collaboration (SINAPSE), Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Karen Horsburgh
- Centre for Neuroregeneration, Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
91
|
At the interface of sensory and motor dysfunctions and Alzheimer's disease. Alzheimers Dement 2015; 11:70-98. [PMID: 25022540 PMCID: PMC4287457 DOI: 10.1016/j.jalz.2014.04.514] [Citation(s) in RCA: 379] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 03/13/2014] [Accepted: 04/09/2014] [Indexed: 11/21/2022]
Abstract
Recent evidence indicates that sensory and motor changes may precede the cognitive symptoms of Alzheimer's disease (AD) by several years and may signify increased risk of developing AD. Traditionally, sensory and motor dysfunctions in aging and AD have been studied separately. To ascertain the evidence supporting the relationship between age-related changes in sensory and motor systems and the development of AD and to facilitate communication between several disciplines, the National Institute on Aging held an exploratory workshop titled "Sensory and Motor Dysfunctions in Aging and AD." The scientific sessions of the workshop focused on age-related and neuropathologic changes in the olfactory, visual, auditory, and motor systems, followed by extensive discussion and hypothesis generation related to the possible links among sensory, cognitive, and motor domains in aging and AD. Based on the data presented and discussed at this workshop, it is clear that sensory and motor regions of the central nervous system are affected by AD pathology and that interventions targeting amelioration of sensory-motor deficits in AD may enhance patient function as AD progresses.
Collapse
|
92
|
Jellinger KA. Pathogenesis and treatment of vascular cognitive impairment. Neurodegener Dis Manag 2014; 4:471-90. [DOI: 10.2217/nmt.14.37] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
SUMMARY Vascular cognitive impairment (VCI) defines a continuum of disorders ranging from mild cognitive impairment to full-blown dementia, attributable to cerebrovascular causes. Major morphological types – multi-infarct encephalopathy, strategic infarct type, subcortical arteriosclerotic leukoencephalopathy, multilacunar state, postischemic encephalopathy – result from systemic, cardiac and local large or small vessel disease. Cognitive decline is commonly caused by widespread small cerebrovascular lesions (CVLs) affecting regions/networks essential for cognition, memory and behavior. CVLs often coexist with Alzheimer-type and other pathologies, which interact in promoting dementia, but in many nondemented elderly individuals, mixed brain pathologies are also present. Due to the high variability of CVLs, no validated clinical and neuropathological criteria for VCI are available. Cholinesterase inhibitors and memantine produce small cognitive improvement but without essential effect. Antihypertensive treatment, cardiovascular control and lifestyle modifications reducing vascular risk factors are essential. Given its growing health, social and economic burden, prevention and treatment of VCI are a major challenge of neuroscience.
Collapse
|
93
|
Al-Mashhadi S, Simpson JE, Heath PR, Dickman M, Forster G, Matthews FE, Brayne C, Ince PG, Wharton SB. Oxidative Glial Cell Damage Associated with White Matter Lesions in the Aging Human Brain. Brain Pathol 2014; 25:565-74. [PMID: 25311358 PMCID: PMC4861214 DOI: 10.1111/bpa.12216] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 10/07/2014] [Indexed: 01/04/2023] Open
Abstract
White matter lesions (WML) are common in brain aging and are associated with dementia. We aimed to investigate whether oxidative DNA damage and occur in WML and in apparently normal white matter in cases with lesions. Tissue from WML and control white matter from brains with lesions (controls lesional) and without lesions (controls non‐lesional) were obtained, using post‐mortem magnetic resonance imaging‐guided sampling, from the Medical Research Council Cognitive Function and Ageing Study. Oxidative damage was assessed by immunohistochemistry to 8‐hydroxy‐2′‐deoxoguanosine (8‐OHdG) and Western blotting for malondialdehyde. DNA response was assessed by phosphorylated histone H2AX (γH2AX), p53, senescence markers and by quantitative Reverse transcription polymerase chain reaction (RT‐PCR) panel for candidate DNA damage‐associated genes. 8‐OHdG was expressed in glia and endothelium, with increased expression in both WML and controls lesional compared with controls non‐lesional (P < 0.001). γH2Ax showed a similar, although attenuated difference among groups (P = 0.03). Expression of senescence‐associated β‐galactosidase and p16 suggested induction of senescence mechanisms in glia. Oxidative DNA damage and a DNA damage response are features of WML pathogenesis and suggest candidate mechanisms for glial dysfunction. Their expression in apparently normal white matter in cases with WML suggests that white matter dysfunction is not restricted to lesions. The role of this field‐effect lesion pathogenesis and cognitive impairment are areas to be defined.
Collapse
Affiliation(s)
- Sufana Al-Mashhadi
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK.,King Fahad Medical City, Riyadh, Saudi Arabia
| | - Julie E Simpson
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Paul R Heath
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Mark Dickman
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | - Gillian Forster
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Fiona E Matthews
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Carol Brayne
- Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Paul G Ince
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | | |
Collapse
|
94
|
Abstract
This report discusses the public health impact of Alzheimer's disease (AD), including incidence and prevalence, mortality rates, costs of care, and overall effect on caregivers and society. It also examines the impact of AD on women compared with men. An estimated 5.2 million Americans have AD. Approximately 200,000 people younger than 65 years with AD comprise the younger onset AD population; 5 million are age 65 years or older. By mid-century, fueled in large part by the baby boom generation, the number of people living with AD in the United States is projected to grow by about 9 million. Today, someone in the country develops AD every 67 seconds. By 2050, one new case of AD is expected to develop every 33 seconds, or nearly a million new cases per year, and the total estimated prevalence is expected to be 13.8 million. In 2010, official death certificates recorded 83,494 deaths from AD, making AD the sixth leading cause of death in the United States and the fifth leading cause of death in Americans aged 65 years or older. Between 2000 and 2010, the proportion of deaths resulting from heart disease, stroke, and prostate cancer decreased 16%, 23%, and 8%, respectively, whereas the proportion resulting from AD increased 68%. The actual number of deaths to which AD contributes (or deaths with AD) is likely much larger than the number of deaths from AD recorded on death certificates. In 2014, an estimated 700,000 older Americans will die with AD, and many of them will die from complications caused by AD. In 2013, more than 15 million family members and other unpaid caregivers provided an estimated 17.7 billion hours of care to people with AD and other dementias, a contribution valued at more than $220 billion. Average per-person Medicare payments for services to beneficiaries aged 65 years and older with AD and other dementias are more than two and a half times as great as payments for all beneficiaries without these conditions, and Medicaid payments are 19 times as great. Total payments in 2014 for health care, long-term care, and hospice services for people aged 65 years and older with dementia are expected to be $214 billion. AD takes a stronger toll on women than men. More women than men develop the disease, and women are more likely than men to be informal caregivers for someone with AD or another dementia. As caregiving responsibilities become more time consuming and burdensome or extend for prolonged durations, women assume an even greater share of the caregiving burden. For every man who spends 21 to more than 60 hours per week as a caregiver, there are 2.1 women. For every man who lives with the care recipient and provides around-the-clock care, there are 2.5 women. In addition, for every man who has provided caregiving assistance for more than 5 years, there are 2.3 women.
Collapse
|
95
|
Attems J, Jellinger KA. The overlap between vascular disease and Alzheimer's disease--lessons from pathology. BMC Med 2014; 12:206. [PMID: 25385447 PMCID: PMC4226890 DOI: 10.1186/s12916-014-0206-2] [Citation(s) in RCA: 478] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 10/07/2014] [Indexed: 12/15/2022] Open
Abstract
Recent epidemiological and clinico-pathological data indicate considerable overlap between cerebrovascular disease (CVD) and Alzheimer's disease (AD) and suggest additive or synergistic effects of both pathologies on cognitive decline. The most frequent vascular pathologies in the aging brain and in AD are cerebral amyloid angiopathy and small vessel disease. Up to 84% of aged subjects show morphological substrates of CVD in addition to AD pathology. AD brains with minor CVD, similar to pure vascular dementia, show subcortical vascular lesions in about two-thirds, while in mixed type dementia (AD plus vascular dementia), multiple larger infarcts are more frequent. Small infarcts in patients with full-blown AD have no impact on cognitive decline but are overwhelmed by the severity of Alzheimer pathology, while in early stages of AD, cerebrovascular lesions may influence and promote cognitive impairment, lowering the threshold for clinically overt dementia. Further studies are warranted to elucidate the many hitherto unanswered questions regarding the overlap between CVD and AD as well as the impact of both CVD and AD pathologies on the development and progression of dementia.
Collapse
Affiliation(s)
- Johannes Attems
- Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK.
| | | |
Collapse
|
96
|
Jellinger KA, Attems J. Challenges of multimorbidity of the aging brain: a critical update. J Neural Transm (Vienna) 2014; 122:505-21. [DOI: 10.1007/s00702-014-1288-x] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 07/24/2014] [Indexed: 12/11/2022]
|
97
|
Vascular and parenchymal lesions along with enhanced neurogenesis characterize the brain of asymptomatic stroke-prone spontaneous hypertensive rats. J Hypertens 2014; 31:1618-28. [PMID: 23666422 DOI: 10.1097/hjh.0b013e3283619d7f] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND AND OBJECTIVES Spontaneously hypertensive stroke-prone rats (SHRSPs) develop hypertension, cerebrovascular abnormalities and a stroke phenotype in association with higher levels of proteinuria. Here, we focus on cerebral abnormalities preceding lesions detectable by MRI. METHODS Longitudinal assessment of brain histology was performed in salt-loaded male SHRSPs (n = 26) and Wistar-Kyoto (WKY) normotensive control animals (n = 27). Groups of rats were sacrificed at different time points: Time 0, before the salt diet administration; Time 1, when proteinuria achieved 40 mg/day; Time 2, when proteinuria exceeded 100 mg/day. RESULTS At Time 0, no brain lesions were observed. At Time 1, changes of the cortical penetrating arteries, vasogenic oedema, lacunae and focal cell loss appeared in SHRSPs and worsened at Time 2, although no lesions were yet detected by MRI. Staining for proliferation markers revealed a significant boost of cellular mitosis in the subventricular zone (SVZ) of SHRSPs. Moreover, we observed higher immunopositivity for nestin, glial fibrillary acidic protein and doublecortin (markers for neural stem cells, astrocytes and immature neurons, respectively). At Time 2, apoptotic caspase-3 as well as 4-hydroxynonenal-positive neurons were associated to decreased nestin and doublecortin staining. High expression levels of glial fibrillary acidic protein were maintained in the SVZ. No comparative alterations and SVZ activation were recorded in WKYs. CONCLUSION Appearance of vascular changes in SHRSPs, before any MRI-detectable brain lesion, is coupled to active neural proliferation in the SVZ. With disease progression, only newborn astrocytes can survive, likely because of the neurotoxicity triggered by brain oedema and oxidative stress.
Collapse
|
98
|
Abstract
Several recent studies demonstrate associations between cardiovascular disease and its risk factors and the incidence of Alzheimer's disease. This review will examine the evidence for these associations and possible pathogenetic pathways. Clinical relevance and implications of these associations will also be discussed.
Collapse
Affiliation(s)
- Caterina Rosano
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | | |
Collapse
|
99
|
Hatano Y, Narumoto J, Shibata K, Matsuoka T, Taniguchi S, Hata Y, Yamada K, Yaku H, Fukui K. White-matter hyperintensities predict delirium after cardiac surgery. Am J Geriatr Psychiatry 2013; 21:938-45. [PMID: 24029014 DOI: 10.1016/j.jagp.2013.01.061] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Revised: 05/31/2012] [Accepted: 06/27/2012] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Postoperative delirium is a common psychiatric disorder among patients who undergo cardiac surgery. Although several studies have investigated risk factors for delirium after cardiac surgery, the association between delirium and cerebral white-matter hyperintensities (WMH) on magnetic resonance (MR) imaging has not been previously studied. The aim of this study was to identify general risk factors for delirium, as well as to examine the specific relationship between WMH and delirium. DESIGN Retrospective chart review. SETTING University hospital. PARTICIPANTS A total of 130 patients who underwent cardiac surgery. MEASUREMENTS Variables recorded included patient demographics, comorbidities, mental health, laboratory data, surgical information, and cerebrovascular disease. The presence of WMH was assessed using MR images. Two groups of patients were compared (patients with and without delirium) using both univariate and multiple logistic analyses. RESULTS Delirium occurred in 18 patients (13.8%) and patients with delirium were significantly older than patients who did not develop delirium. The prevalence of severe WMH (Fazekas score = 3) was significantly higher in patients with delirium. Three independent predictors of delirium were identified: abnormal creatinine (odds ratio [OR]: 4.5; 95% confidence interval [CI]: 1.4-13.9), severe WMH (OR: 3.9; 95% CI: 1.2-12.5), and duration of surgery (OR: 1.4; 95% CI: 1.0-1.8). CONCLUSIONS The results of this study suggest that white-matter abnormality is one of the most important risk factors for development of delirium after cardiac surgery. These factors can be used for prediction and prevention of delirium following cardiac surgery.
Collapse
Affiliation(s)
- Yutaka Hatano
- Department of Psychiatry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Minn YK, Suk SH, Park H, Cheong JS, Yang H, Lee S, Do SY, Kang JS. Tooth loss is associated with brain white matter change and silent infarction among adults without dementia and stroke. J Korean Med Sci 2013; 28:929-33. [PMID: 23772160 PMCID: PMC3678012 DOI: 10.3346/jkms.2013.28.6.929] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 04/16/2013] [Indexed: 12/28/2022] Open
Abstract
Periodontal disease is a predictor of stroke and cognitive impairment. The association between the number of lost teeth (an indicator of periodontal disease) and silent infarcts and cerebral white matter changes on brain CT was investigated in community-dwelling adults without dementia or stroke. Dental examination and CT were performed in 438 stroke- and dementia-free subjects older than 50 yr (mean age, 63 ± 7.9 yr), who were recruited for an early health check-up program as part of the Prevention of Stroke and Dementia (PRESENT) project between 2009 and 2010. In unadjusted analyses, the odds ratio (OR) for silent cerebral infarcts and cerebral white matter changes for subjects with 6-10 and > 10 lost teeth was 2.3 (95% CI, 1.38-4.39; P = 0.006) and 4.2 (95% CI, 1.57-5.64; P < 0.001), respectively, as compared to subjects with 0-5 lost teeth. After adjustment for age, education, hypertension, diabetes mellitus, hyperlipidemia, and smoking, the ORs were 1.7 (95% CI, 1.08-3.69; P = 0.12) and 3.9 (95% CI, 1.27-5.02; P < 0.001), respectively. These findings suggest that severe tooth loss may be a predictor of silent cerebral infarcts and cerebral white matter changes in community-dwelling, stroke- and dementia-free adults.
Collapse
Affiliation(s)
- Yang-Ki Minn
- Department of Neurology, Hallym University, Seoul, Korea
| | - Seung-Han Suk
- Department of Neurology, Wonkwang University Sanbon Medical Center, Gunpo, Korea
- Wonkwang University Ansan Municipal Geriatric Hospital and Center for Prevention of Stroke and Dementia, Ansan, Korea
| | - Hyunyoung Park
- Department of Neurology, Wonkwang University, Iksan, Korea
| | | | - Hyunduk Yang
- Department of Neurology, Wonkwang University Sanbon Medical Center, Gunpo, Korea
| | - Sungik Lee
- Department of Neurology, Wonkwang University Sanbon Medical Center, Gunpo, Korea
| | - Seung-Yeon Do
- Division of Health and Nutrition Survey, Korea Centers for Disease Control and Prevention, Cheongwon, Korea
| | - Ji-Sook Kang
- Department of Nursing, Wonkwang University College of Medicine, Iksan, Korea
| |
Collapse
|