1
|
Chen F, Zhao J, Meng F, He F, Ni J, Fu Y. The vascular contribution of apolipoprotein E to Alzheimer's disease. Brain 2024; 147:2946-2965. [PMID: 38748848 DOI: 10.1093/brain/awae156] [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] [Received: 12/24/2023] [Revised: 03/23/2024] [Accepted: 04/21/2024] [Indexed: 09/04/2024] Open
Abstract
Alzheimer's disease, the most prevalent form of dementia, imposes a substantial societal burden. The persistent inadequacy of disease-modifying drugs targeting amyloid plaques and neurofibrillary tangles suggests the contribution of alternative pathogenic mechanisms. A frequently overlooked aspect is cerebrovascular dysfunction, which may manifest early in the progression of Alzheimer's disease pathology. Mounting evidence underscores the pivotal role of the apolipoprotein E gene, particularly the apolipoprotein ε4 allele as the strongest genetic risk factor for late-onset Alzheimer's disease, in the cerebrovascular pathology associated with Alzheimer's disease. In this review, we examine the evidence elucidating the cerebrovascular impact of both central and peripheral apolipoprotein E on the pathogenesis of Alzheimer's disease. We present a novel three-hit hypothesis, outlining potential mechanisms that shed light on the intricate relationship among different pathogenic events. Finally, we discuss prospective therapeutics targeting the cerebrovascular pathology associated with apolipoprotein E and explore their implications for future research endeavours.
Collapse
Affiliation(s)
- Feng Chen
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jing Zhao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Fanxia Meng
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Fangping He
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jie Ni
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuan Fu
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
2
|
Ma X, Gao H, Wu Y, Zhu X, Wu S, Lin L. Investigating Modifiable Risk Factors Across Dementia Subtypes: Insights from the UK Biobank. Biomedicines 2024; 12:1967. [PMID: 39335481 PMCID: PMC11428917 DOI: 10.3390/biomedicines12091967] [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: 07/22/2024] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
This study investigates the relationship between modifiable risk factors and dementia subtypes using data from 460,799 participants in the UK Biobank. Utilizing univariate Cox proportional hazards regression models, we examined the associations between 83 modifiable risk factors and the risks of all-cause dementia (ACD), Alzheimer's disease (AD), and vascular dementia (VD). Composite scores for different domains were generated by aggregating risk factors associated with ACD, AD, and VD, respectively, and their joint associations were assessed in multivariable Cox models. Additionally, population attributable fractions (PAF) were utilized to estimate the potential impact of eliminating adverse characteristics of the risk domains. Our findings revealed that an unfavorable medical history significantly increased the risk of ACD, AD, and VD (hazard ratios (HR) = 1.88, 95% confidence interval (95% CI): 1.74-2.03, p < 0.001; HR = 1.80, 95% CI: 1.54-2.10, p < 0.001; HR = 2.39, 95% CI: 2.10-2.71, p < 0.001, respectively). Blood markers (PAF = 12.1%; 17.4%) emerged as the most important risk domain for preventing ACD and VD, while psychiatric factors (PAF = 18.3%) were the most important for preventing AD. This study underscores the potential for preventing dementia and its subtypes through targeted interventions for modifiable risk factors. The distinct insights provided by HR and PAF emphasize the importance of considering both the strength of the associations and the population-level impact of dementia prevention strategies. Our research provides valuable guidance for developing effective public health interventions aimed at reducing the burden of dementia, representing a significant advancement in the field.
Collapse
Affiliation(s)
| | | | | | | | | | - Lan Lin
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China; (X.M.); (H.G.); (Y.W.); (X.Z.); (S.W.)
| |
Collapse
|
3
|
Granholm ACE, Englund E, Gilmore A, Head E, Yong WH, Perez SE, Guzman SJ, Hamlett ED, Mufson EJ. Neuropathological findings in Down syndrome, Alzheimer's disease and control patients with and without SARS-COV-2: preliminary findings. Acta Neuropathol 2024; 147:92. [PMID: 38801558 PMCID: PMC11130011 DOI: 10.1007/s00401-024-02743-9] [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] [Received: 02/20/2024] [Revised: 05/11/2024] [Accepted: 05/12/2024] [Indexed: 05/29/2024]
Abstract
The SARS-CoV-2 virus that led to COVID-19 is associated with significant and long-lasting neurologic symptoms in many patients, with an increased mortality risk for people with Alzheimer's disease (AD) and/or Down syndrome (DS). However, few studies have evaluated the neuropathological and inflammatory sequelae in postmortem brain tissue obtained from AD and people with DS with severe SARS-CoV-2 infections. We examined tau, beta-amyloid (Aβ), inflammatory markers and SARS-CoV-2 nucleoprotein in DS, AD, and healthy non-demented controls with COVID-19 and compared with non-infected brain tissue from each disease group (total n = 24). A nested ANOVA was used to determine regional effects of the COVID-19 infection on arborization of astrocytes (Sholl analysis) and percent-stained area of Iba-1 and TMEM 119. SARS-CoV-2 antibodies labeled neurons and glial cells in the frontal cortex of all subjects with COVID-19, and in the hippocampus of two of the three DS COVID-19 cases. SARS-CoV-2-related alterations were observed in peri-vascular astrocytes and microglial cells in the gray matter of the frontal cortex, hippocampus, and para-hippocampal gyrus. Bright field microscopy revealed scattered intracellular and diffuse extracellular Aβ deposits in the hippocampus of controls with confirmed SARS-CoV-2 infections. Overall, the present preliminary findings suggest that SARS-CoV-2 infections induce abnormal inflammatory responses in Down syndrome.
Collapse
Affiliation(s)
- Ann-Charlotte E Granholm
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Research Complex II, Aurora, CO, USA.
| | - Elisabet Englund
- Division of Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Anah Gilmore
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Research Complex II, Aurora, CO, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA, USA
- Department of Neurology, University of California Irvine, Irvine, CA, USA
| | - William H Yong
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA, USA
| | - Sylvia E Perez
- Department of Translational Neuroscience and Neurology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Samuel J Guzman
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Eric D Hamlett
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Elliott J Mufson
- Department of Translational Neuroscience and Neurology, Barrow Neurological Institute, Phoenix, AZ, USA
| |
Collapse
|
4
|
Cao F, Yang F, Li J, Guo W, Zhang C, Gao F, Sun X, Zhou Y, Zhang W. The relationship between diabetes and the dementia risk: a meta-analysis. Diabetol Metab Syndr 2024; 16:101. [PMID: 38745237 PMCID: PMC11092065 DOI: 10.1186/s13098-024-01346-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 05/04/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND The link between diabetes and dementia risk is not well understood. This study evaluates the factors linking diabetes to dementia onset, providing guidance for preventing dementia in diabetic patients. METHODS This analysis utilized databases such as PubMed, Embase, Web of Science, and the Cochrane Library to review literature from January 31, 2012, to March 5, 2023. Articles were rigorously assessed using specific inclusion and exclusion criteria. The Newcastle-Ottawa Scale (NOS) was used to evaluate the quality of the studies. Data analysis was performed with STATA 15.0. RESULTS The study analyzed 15 articles, covering 10,103,868 patients, with 8,821,516 diagnosed with diabetes. The meta-analysis reveals a substantial association between diabetes and an increased risk of dementia [RR: 1.59, 95%CI (1.40-1.80), P < 0.01, I²=96.4%]. A diabetes duration of less than five years is linked to a higher dementia risk [RR: 1.29, 95%CI (1.20-1.39), P < 0.01, I²=92.6%]. Additionally, hypoglycemia significantly raises dementia risk [RR: 1.56, 95%CI (1.13-2.16), P < 0.01, I²=51.5%]. Analyses of blood sugar control, glycated hemoglobin, and fasting blood sugar indicated no significant effects on the onset of dementia. CONCLUSION Diabetes notably increases dementia risk, particularly where diabetes duration is under five years or hypoglycemia is present. REGISTRATION The research protocol was registered with PROSPERO and assigned the registration number CRD42023394942.
Collapse
Affiliation(s)
- Fang Cao
- School of Basic Medical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Fushuang Yang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Jian Li
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Wei Guo
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Chongheng Zhang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Fa Gao
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Xinxin Sun
- Department of Nutrition, Chinese People's Armed Police Force Medical Characteristic Center, Tianjin, 300162, China
| | - Yi Zhou
- Department of Geriatrics, Baotou Mengshi Hospital of Traditional Chinese Medicine, Baotou, 014000, China
| | - Wenfeng Zhang
- School of Basic Medical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China.
| |
Collapse
|
5
|
Morgan AE, Mc Auley MT. Vascular dementia: From pathobiology to emerging perspectives. Ageing Res Rev 2024; 96:102278. [PMID: 38513772 DOI: 10.1016/j.arr.2024.102278] [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: 01/26/2024] [Revised: 03/16/2024] [Accepted: 03/17/2024] [Indexed: 03/23/2024]
Abstract
Vascular dementia (VaD) is the second most common type of dementia. VaD is synonymous with ageing, and its symptoms place a significant burden on the health and wellbeing of older people. Despite the identification of a substantial number of risk factors for VaD, the pathological mechanisms underpinning this disease remain to be fully elucidated. Consequently, a biogerontological imperative exists to highlight the modifiable lifestyle factors which can mitigate against the risk of developing VaD. This review will critically examine some of the factors which have been revealed to modulate VaD risk. The survey commences by providing an overview of the putative mechanisms which are associated with the pathobiology of VaD. Next, the factors which influence the risk of developing VaD are examined. Finally, emerging treatment avenues including epigenetics, the gut microbiome, and pro-longevity pharmaceuticals are discussed. By drawing this key evidence together, it is our hope that it can be used to inform future experimental investigations in this field.
Collapse
Affiliation(s)
- Amy Elizabeth Morgan
- School of Health and Sports Sciences, Hope Park, Liverpool Hope University, Liverpool L16 9JD, United Kingdom.
| | - Mark Tomás Mc Auley
- School of Science, Engineering and Environment, University of Salford Manchester, Salford M5 4NT, United Kingdom
| |
Collapse
|
6
|
Sah RP, Vidya CS, Pereira P, Jayaram S, Yadav AK, Sujatha P. Elevated Homocysteine Level and Brain Atrophy Changes as Markers to Screen the Alzheimer Disease: Case Series. Ann Geriatr Med Res 2024; 28:116-120. [PMID: 38105011 PMCID: PMC10982442 DOI: 10.4235/agmr.23.0135] [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: 08/19/2023] [Revised: 11/09/2023] [Accepted: 12/08/2023] [Indexed: 12/19/2023] Open
Abstract
Alzheimer disease (AD) is the most common cause of dementia worldwide. Its clinical manifestations include a progressive loss of memory and other cognitive domains, as well as brain atrophy. An elevated homocysteine level (>15 µmol/L), known as hyperhomocysteinemia, is also an attributing risk factor for AD, vascular pathologies, and brain atrophy. Neuroimaging studies including T2-weighted magnetic resonance imaging scans revealed white matter hyperintensities in the periventricular and deep white matter, enlarged ventricles, widened sulci, and decreased white matter mass, which are features of aging, as well as cerebrovascular changes. This case series investigated changes in biochemical marker levels including serum homocysteine, folate, and vitamin B12, and the degree of atrophic variations in cortical-subcortical white matter in AD. The present study hypothesized that serum homocysteine levels might be used as a surrogate marker to screen for AD at an earlier stage.
Collapse
Affiliation(s)
- Ram Prakash Sah
- Department of Anatomy, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, India
| | - C. S. Vidya
- Department of Anatomy, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, India
| | - Pratibha Pereira
- Department of Geriatric Medicine, JSS Hospital, JSS Academy of Higher Education and Research, Mysore, India
| | - Shubha Jayaram
- Department of Biochemistry, Mysore Medical College and Research Institute, Mysore, India
| | - Anshu Kumar Yadav
- Department of Biochemistry and Medical Genetics, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, India
| | - P. Sujatha
- Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, India
| |
Collapse
|
7
|
Blood pressure targets for prevention of cognitive decline in patients with diabetes and hypertension: Design of the Blood Pressure Control Target in Diabetes (BPROAD) Cognitive Study. J Diabetes 2023; 15:1041-1047. [PMID: 37737064 PMCID: PMC10755604 DOI: 10.1111/1753-0407.13412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 11/12/2021] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND Both hypertension and diabetes are risk factors of dementia. Proper management of blood pressure (BP) and blood glucose is critical in delaying cognitive decline in the elderly. However, little is known regarding the optimal BP target in type 2 diabetes (T2DM) for the management of cognitive decline. METHODS The Blood Pressure Control Target in Diabetes (BPROAD) study is a nationwide, multicenter, randomized controlled trial that will enroll 12 702 T2DM patients with elevated systolic BP and increased cardiovascular risk from approximately 150 study centers across mainland China to undergo randomly antihypertensive treatment achieving systolic BP <120 mm Hg or systolic BP <140 mm Hg for up to 5 years. All BPROAD participants will take part in the BPROAD Cognitive Study for the assessment of cognitive function at baseline and annual visits by blinded outcome assessors to determine whether intensive BP treatment reduces risk of dementia and mild cognitive impairment (MCI) compared with standard BP treatment in patients with T2DM. In addition, approximately 1000 BPROAD participants will be enrolled in the magnetic resonance imaging (MRI) substudy to receive brain MRI at baseline and at closeout. The primary outcome of BPROAD Cognitive Study is a composite of all-cause dementia and MCI. CONCLUSIONS The BPROAD Cognitive Study will provide crucial clinical trial data on the possible benefit of an intensive systolic BP lowering strategy in reducing dementia and MCI in patients with T2DM.
Collapse
|
8
|
Pandics T, Major D, Fazekas-Pongor V, Szarvas Z, Peterfi A, Mukli P, Gulej R, Ungvari A, Fekete M, Tompa A, Tarantini S, Yabluchanskiy A, Conley S, Csiszar A, Tabak AG, Benyo Z, Adany R, Ungvari Z. Exposome and unhealthy aging: environmental drivers from air pollution to occupational exposures. GeroScience 2023; 45:3381-3408. [PMID: 37688657 PMCID: PMC10643494 DOI: 10.1007/s11357-023-00913-3] [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] [Received: 06/22/2023] [Accepted: 08/14/2023] [Indexed: 09/11/2023] Open
Abstract
The aging population worldwide is facing a significant increase in age-related non-communicable diseases, including cardiovascular and brain pathologies. This comprehensive review paper delves into the impact of the exposome, which encompasses the totality of environmental exposures, on unhealthy aging. It explores how environmental factors contribute to the acceleration of aging processes, increase biological age, and facilitate the development and progression of a wide range of age-associated diseases. The impact of environmental factors on cognitive health and the development of chronic age-related diseases affecting the cardiovascular system and central nervous system is discussed, with a specific focus on Alzheimer's disease, Parkinson's disease, stroke, small vessel disease, and vascular cognitive impairment (VCI). Aging is a major risk factor for these diseases. Their pathogenesis involves cellular and molecular mechanisms of aging such as increased oxidative stress, impaired mitochondrial function, DNA damage, and inflammation and is influenced by environmental factors. Environmental toxicants, including ambient particulate matter, pesticides, heavy metals, and organic solvents, have been identified as significant contributors to cardiovascular and brain aging disorders. These toxicants can inflict both macro- and microvascular damage and many of them can also cross the blood-brain barrier, inducing neurotoxic effects, neuroinflammation, and neuronal dysfunction. In conclusion, environmental factors play a critical role in modulating cardiovascular and brain aging. A deeper understanding of how environmental toxicants exacerbate aging processes and contribute to the pathogenesis of neurodegenerative diseases, VCI, and dementia is crucial for the development of preventive strategies and interventions to promote cardiovascular, cerebrovascular, and brain health. By mitigating exposure to harmful environmental factors and promoting healthy aging, we can strive to reduce the burden of age-related cardiovascular and brain pathologies in the aging population.
Collapse
Affiliation(s)
- Tamas Pandics
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Department of Public Health Laboratory, National Public Health Centre, Budapest, Hungary
- Department of Public Health Siences, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - David Major
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Vince Fazekas-Pongor
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Zsofia Szarvas
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Peterfi
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Mukli
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Ungvari
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Monika Fekete
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Anna Tompa
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Shannon Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Adam G Tabak
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- UCL Brain Sciences, University College London, London, UK
- Department of Internal Medicine and Oncology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Zoltan Benyo
- Department of Translational Medicine, Semmelweis University, Budapest, Hungary
- Eötvös Loránd Research Network and Semmelweis University (ELKH-SE) Cerebrovascular and Neurocognitive Disorders Research Group, Budapest, H-1052, Hungary
| | - Roza Adany
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- ELKH-DE Public Health Research Group, Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary
- Epidemiology and Surveillance Centre, Semmelweis University, 1085, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA.
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
9
|
Pedersen MK, Eriksson R, Reguant R, Collin C, Pedersen HK, Sørup FKH, Simon C, Birch AM, Larsen M, Nielsen AP, Belling K, Brunak S. A unidirectional mapping of ICD-8 to ICD-10 codes, for harmonized longitudinal analysis of diseases. Eur J Epidemiol 2023; 38:1043-1052. [PMID: 37555907 PMCID: PMC10570238 DOI: 10.1007/s10654-023-01027-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/22/2023] [Indexed: 08/10/2023]
Abstract
Periodic revisions of the international classification of diseases (ICD) ensure that the classification reflects new practices and knowledge; however, this complicates retrospective research as diagnoses are coded in different versions. For longitudinal disease trajectory studies, a crosswalk is an essential tool and a comprehensive mapping between ICD-8 and ICD-10 has until now been lacking. In this study, we map all ICD-8 morbidity codes to ICD-10 in the expanded Danish ICD version. We mapped ICD-8 codes to ICD-10, using a many-to-one system inspired by general equivalence mappings such that each ICD-8 code maps to a single ICD-10 code. Each ICD-8 code was manually and unidirectionally mapped to a single ICD-10 code based on medical setting and context. Each match was assigned a score (1 of 4 levels) reflecting the quality of the match and, if applicable, a "flag" signalling choices made in the mapping. We provide the first complete mapping of the 8596 ICD-8 morbidity codes to ICD-10 codes. All Danish ICD-8 codes representing diseases were mapped and 5106 (59.4%) achieved the highest consistency score. Only 334 (3.9%) of the ICD-8 codes received the lowest mapping consistency score. The mapping provides a scaffold for translation of ICD-8 to ICD-10, which enable longitudinal disease studies back to and 1969 in Denmark and to 1965 internationally with further adaption.
Collapse
Affiliation(s)
- Mette Krogh Pedersen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Robert Eriksson
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, 2200, Copenhagen, Denmark
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Division of Infectious Diseases, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Roc Reguant
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, 2200, Copenhagen, Denmark
- Australian E-Health Research Centre, Commonwealth Scientific and Industrial Research Organisation, Sydney, Australia
| | - Catherine Collin
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Helle Krogh Pedersen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, 2200, Copenhagen, Denmark
| | | | - Christian Simon
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Anna Marie Birch
- Department of Obstetrics and Gynaecology, Holbæk Hospital, Holbæk, Denmark
| | - Michael Larsen
- Department of Ophthalmology, Rigshospitalet-Glostrup, Copenhagen University Hospital, Glostrup, Denmark
| | - Anna Pors Nielsen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Kirstine Belling
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, 2200, Copenhagen, Denmark.
| |
Collapse
|
10
|
Juul Rasmussen I, Frikke-Schmidt R. Modifiable cardiovascular risk factors and genetics for targeted prevention of dementia. Eur Heart J 2023; 44:2526-2543. [PMID: 37224508 PMCID: PMC10481783 DOI: 10.1093/eurheartj/ehad293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 02/22/2023] [Accepted: 05/04/2023] [Indexed: 05/26/2023] Open
Abstract
Dementia is a major global challenge for health and social care in the 21st century. A third of individuals >65 years of age die with dementia, and worldwide incidence numbers are projected to be higher than 150 million by 2050. Dementia is, however, not an inevitable consequence of old age; 40% of dementia may theoretically be preventable. Alzheimer's disease (AD) accounts for approximately two-thirds of dementia cases and the major pathological hallmark of AD is accumulation of amyloid-β. Nevertheless, the exact pathological mechanisms of AD remain unknown. Cardiovascular disease and dementia share several risk factors and dementia often coexists with cerebrovascular disease. In a public health perspective, prevention is crucial, and it is suggested that a 10% reduction in prevalence of cardiovascular risk factors could prevent more than nine million dementia cases worldwide by 2050. Yet this assumes causality between cardiovascular risk factors and dementia and adherence to the interventions over decades for a large number of individuals. Using genome-wide association studies, the entire genome can be scanned for disease/trait associated loci in a hypothesis-free manner, and the compiled genetic information is not only useful for pinpointing novel pathogenic pathways but also for risk assessments. This enables identification of individuals at high risk, who likely will benefit the most from a targeted intervention. Further optimization of the risk stratification can be done by adding cardiovascular risk factors. Additional studies are, however, highly needed to elucidate dementia pathogenesis and potential shared causal risk factors between cardiovascular disease and dementia.
Collapse
Affiliation(s)
- Ida Juul Rasmussen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
11
|
Dybjer E, Kumar A, Nägga K, Engström G, Mattsson-Carlgren N, Nilsson PM, Melander O, Hansson O. Polygenic risk of type 2 diabetes is associated with incident vascular dementia: a prospective cohort study. Brain Commun 2023; 5:fcad054. [PMID: 37091584 PMCID: PMC10118265 DOI: 10.1093/braincomms/fcad054] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/28/2022] [Accepted: 03/02/2023] [Indexed: 03/09/2023] Open
Abstract
Type 2 diabetes and dementia are associated, but it is unclear whether the two diseases have common genetic risk markers that could partly explain their association. It is also unclear whether the association between the two diseases is of a causal nature. Furthermore, few studies on diabetes and dementia have validated dementia end-points with high diagnostic precision. We tested associations between polygenic risk scores for type 2 diabetes, fasting glucose, fasting insulin and haemoglobin A1c as exposure variables and dementia as outcome variables in 29 139 adults (mean age 55) followed for 20-23 years. Dementia diagnoses were validated by physicians through data from medical records, neuroimaging and biomarkers in cerebrospinal fluid. The dementia end-points included all-cause dementia, mixed dementia, Alzheimer's disease and vascular dementia. We also tested causal associations between type 2 diabetes and dementia through two-sample Mendelian randomization analyses. Seven different polygenic risk scores including single-nucleotide polymorphisms with different significance thresholds for type 2 diabetes were tested. A polygenic risk score including 4891 single-nucleotide polymorphisms with a P-value of <5e-04 showed the strongest association with different outcomes, including all-cause dementia (hazard ratio 1.11; Bonferroni corrected P = 3.6e-03), mixed dementia (hazard ratio 1.18; Bonferroni corrected P = 3.3e-04) and vascular dementia cases (hazard ratio 1.28; Bonferroni corrected P = 9.6e-05). The associations were stronger for non-carriers of the Alzheimer's disease risk gene APOE ε4. There was, however, no significant association between polygenic risk scores for type 2 diabetes and Alzheimer's disease. Furthermore, two-sample Mendelian randomization analyses could not confirm a causal link between genetic risk markers of type 2 diabetes and dementia outcomes. In conclusion, polygenic risk of type 2 diabetes is associated with an increased risk of dementia, in particular vascular dementia. The findings imply that certain people with type 2 diabetes may, due to their genetic background, be more prone to develop diabetes-associated dementia. This knowledge could in the future lead to targeted preventive strategies in clinical practice.
Collapse
Affiliation(s)
- Elin Dybjer
- Department of Clinical Sciences Malmö, Lund University, Jan Waldenströms gata 35, SE-21428 Malmö, Sweden
| | - Atul Kumar
- MultiPark: Multidisciplinary Research focused on Parkinson's disease, Lund University, Box 117, SE-22100 Lund, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Skånes universitetssjukhus, VE Minnessjukdomar, SE-20502 Malmö, Sweden
| | - Katarina Nägga
- Department of Clinical Sciences Malmö, Lund University, Jan Waldenströms gata 35, SE-21428 Malmö, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Skånes universitetssjukhus, VE Minnessjukdomar, SE-20502 Malmö, Sweden
- Department of Acute Internal Medicine and Geriatrics, Linköping University, SE-58183 Linköping, Sweden
| | - Gunnar Engström
- Department of Clinical Sciences Malmö, Lund University, Jan Waldenströms gata 35, SE-21428 Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- MultiPark: Multidisciplinary Research focused on Parkinson's disease, Lund University, Box 117, SE-22100 Lund, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Skånes universitetssjukhus, VE Minnessjukdomar, SE-20502 Malmö, Sweden
- Brain Injury After Cardiac Arrest Research Group, Lund University, Box 117, SE-22100 Lund, Sweden
- WCMM – Wallenberg Centre for Molecular Medicine, Lund University, Sölvegatan 19, BMC D11, SE-22184 Lund, Sweden
| | - Peter M Nilsson
- Department of Clinical Sciences Malmö, Lund University, Jan Waldenströms gata 35, SE-21428 Malmö, Sweden
- EpiHealth: Epidemiology for Health Strategic Research Area, Lund University, SUS Malmö, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden
| | - Olle Melander
- Department of Clinical Sciences Malmö, Lund University, Jan Waldenströms gata 35, SE-21428 Malmö, Sweden
- EpiHealth: Epidemiology for Health Strategic Research Area, Lund University, SUS Malmö, Jan Waldenströms gata 35, SE-20502 Malmö, Sweden
- Department of Emergency and Internal Medicine, Skåne University Hospital, SE-20502 Malmö, Sweden
- EXODIAB: Excellence in Diabetes Research in Sweden, Lund University, Box 117, SE-22100 Lund, Sweden
| | - Oskar Hansson
- MultiPark: Multidisciplinary Research focused on Parkinson's disease, Lund University, Box 117, SE-22100 Lund, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Skånes universitetssjukhus, VE Minnessjukdomar, SE-20502 Malmö, Sweden
| |
Collapse
|
12
|
Javanshiri K, Drakenberg T, Haglund M, Englund E. Sudden cardiac death in synucleinopathies. J Neuropathol Exp Neurol 2023; 82:242-249. [PMID: 36668680 PMCID: PMC9941831 DOI: 10.1093/jnen/nlad001] [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] [Indexed: 01/22/2023] Open
Abstract
The purpose of this study was to investigate the cause of death in subjects with α-synucleinopathies (ASs) and the confirmed presence of cardiac α-synuclein (α-syn), compared to non-AS disorders in a neuropathologically confirmed cohort. In total, 78 neuropathologically confirmed AS cases positive for cardiac α-syn were included in the study. Individuals with other neurocognitive diseases, having no α-syn in the brainstem or above, nor in cardiac nerves, served as controls (n = 53). Data regarding the cause of death, cardiac α-syn, pathological cardiac findings, and cardio- and cerebrovascular disease were assembled from autopsy reports and medical records. In the AS group, there was a significantly higher prevalence of sudden cardiac death ([SCD]; n = 40, 51.3%) compared to the control group (n = 12, 22.6%, p < 0.001). No statistically significant differences between the groups were reported regarding other cardiac conditions on autopsy or regarding cardio- and cerebrovascular disease from the medical records. The most prevalent cause of death in the AS group was SCD, which differed significantly from the control group. This suggests that α-syn deposits in cardiac nerves may cause lethal alterations in cardiac function, warranting further research.
Collapse
Affiliation(s)
- Keivan Javanshiri
- Division of Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Tove Drakenberg
- Division of Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Mattias Haglund
- Division of Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Elisabet Englund
- Division of Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
13
|
Vellecco V, Saviano A, Raucci F, Casillo GM, Mansour AA, Panza E, Mitidieri E, Femminella GD, Ferrara N, Cirino G, Sorrentino R, Iqbal AJ, d'Emmanuele di Villa Bianca R, Bucci M, Maione F. Interleukin-17 (IL-17) triggers systemic inflammation, peripheral vascular dysfunction, and related prothrombotic state in a mouse model of Alzheimer's disease. Pharmacol Res 2023; 187:106595. [PMID: 36470548 DOI: 10.1016/j.phrs.2022.106595] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/22/2022] [Accepted: 11/30/2022] [Indexed: 12/08/2022]
Abstract
Alzheimer's disease (AD) is one of the most prevalent forms of neurodegenerative disorders. Previously, we have shown that in vivo administration of an IL-17 neutralizing antibody (IL-17Ab) rescues amyloid-β-induced neuro-inflammation and memory impairment, demonstrating the pivotal role of IL-17 in AD-derived cognitive deficit. Recently, AD has been recognized as a more intriguing pathology affecting vascular networks and platelet function. However, not much is known about peripheral vascular inflammation and how pro-inflammatory circulating cells/mediators could affect peripheral vessels' function. This study aimed to evaluate whether IL-17Ab treatment could also impact peripheral AD features, such as systemic inflammation, peripheral vascular dysfunction, and related pro-thrombotic state in a non-genetic mouse model of AD. Mice were injected intracerebroventricularly with Aβ1-42 peptide (3 μg/3 μl). To evaluate the systemic/peripheral protective profile of IL-17Ab, we used an intranasal administration of IL-17Ab (1 μg/10 μl) at 5, 12, and 19 days after Aβ1-42 injection. Circulating Th17/Treg cells and related cyto-chemokines, haematological parameters, vascular/endothelial reactivity, platelets and coagulation function in mice were evaluated. IL-17Ab treatment ameliorates the systemic/peripheral inflammation, immunological perturbance, vascular/endothelial impairment and pro-thrombotic state, suggesting a key role for this cytokine in fostering inflammatory processes that characterize the multifaced aspects of AD.
Collapse
Affiliation(s)
- Valentina Vellecco
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Anella Saviano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Federica Raucci
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Gian Marco Casillo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Adel Abo Mansour
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.
| | - Elisabetta Panza
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Emma Mitidieri
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Grazia Daniela Femminella
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy.
| | - Nicola Ferrara
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; Istituti Clinici Scientifici ICS-Maugeri, Telese Terme, BN, Italy.
| | - Giuseppe Cirino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Raffaella Sorrentino
- Department of Molecular Medicine and Medical Biotechnologies, School of Medicine, University of Naples, Federico II, Via Pansini, 5, 80131 Naples, Italy.
| | - Asif Jilani Iqbal
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy; Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| | | | - Mariarosaria Bucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| | - Francesco Maione
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy.
| |
Collapse
|
14
|
Kim SE, Kim HJ, Jang H, Weiner MW, DeCarli C, Na DL, Seo SW. Interaction between Alzheimer's Disease and Cerebral Small Vessel Disease: A Review Focused on Neuroimaging Markers. Int J Mol Sci 2022; 23:10490. [PMID: 36142419 PMCID: PMC9499680 DOI: 10.3390/ijms231810490] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/07/2022] [Accepted: 09/07/2022] [Indexed: 11/26/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by the presence of β-amyloid (Aβ) and tau, and subcortical vascular cognitive impairment (SVCI) is characterized by cerebral small vessel disease (CSVD). They are the most common causes of cognitive impairment in the elderly population. Concurrent CSVD burden is more commonly observed in AD-type dementia than in other neurodegenerative diseases. Recent developments in Aβ and tau positron emission tomography (PET) have enabled the investigation of the relationship between AD biomarkers and CSVD in vivo. In this review, we focus on the interaction between AD and CSVD markers and the clinical effects of these two markers based on molecular imaging studies. First, we cover the frequency of AD imaging markers, including Aβ and tau, in patients with SVCI. Second, we discuss the relationship between AD and CSVD markers and the potential distinct pathobiology of AD markers in SVCI compared to AD-type dementia. Next, we discuss the clinical effects of AD and CSVD markers in SVCI, and hemorrhagic markers in cerebral amyloid angiopathy. Finally, this review provides both the current challenges and future perspectives for SVCI.
Collapse
Affiliation(s)
- Si Eun Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Neuroscience Center, Samsung Medical Center, Seoul 06351, Korea
- Samsung Alzheimer Research Center, Samsung Medical Center, Seoul 06351, Korea
- Department of Neurology, Inje University College of Medicine, Haeundae Paik Hospital, Busan 48108, Korea
| | - Hee Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Neuroscience Center, Samsung Medical Center, Seoul 06351, Korea
- Samsung Alzheimer Research Center, Samsung Medical Center, Seoul 06351, Korea
| | - Hyemin Jang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Neuroscience Center, Samsung Medical Center, Seoul 06351, Korea
- Samsung Alzheimer Research Center, Samsung Medical Center, Seoul 06351, Korea
| | - Michael W. Weiner
- Center for Imaging of Neurodegenerative Diseases, University of California, San Francisco, CA 94121, USA
| | - Charles DeCarli
- Department of Neurology and Center for Neuroscience, University of California, Davis, CA 95616, USA
| | - Duk L. Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Neuroscience Center, Samsung Medical Center, Seoul 06351, Korea
- Samsung Alzheimer Research Center, Samsung Medical Center, Seoul 06351, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul 06355, Korea
- Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul 06351, Korea
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
- Neuroscience Center, Samsung Medical Center, Seoul 06351, Korea
- Samsung Alzheimer Research Center, Samsung Medical Center, Seoul 06351, Korea
- Department of Clinical Research Design and Evaluation, SAIHST, Sungkyunkwan University, Seoul 06355, Korea
- Center for Clinical Epidemiology, Samsung Medical Center, Seoul 06351, Korea
| |
Collapse
|
15
|
Cao Z, Mai Y, Fang W, Lei M, Luo Y, Zhao L, Liao W, Yu Q, Xu J, Ruan Y, Xiao S, Mok VCT, Shi L, Liu J. The Correlation Between White Matter Hyperintensity Burden and Regional Brain Volumetry in Patients With Alzheimer's Disease. Front Hum Neurosci 2022; 16:760360. [PMID: 35774484 PMCID: PMC9237397 DOI: 10.3389/fnhum.2022.760360] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
Background White matter hyperintensities (WMHs) and regional brain lobe atrophy coexist in the brain of patients with Alzheimer's disease (AD), but the association between them in patients with AD still lacks comprehensive investigation and solid imaging data support. Objective We explored whether WMHs can promote the pathological process of AD by aggravating atrophy in specific brain regions and tried to explain the regional specificity of these relationships. Methods A sample of 240 adults including 180 normal controls (NCs) and 80 cases with AD were drawn from the ADNI database. T1-weighted magnetic resonance imaging (MRI) and T2-weighted fluid-attenuated MRI of the participants were downloaded and were analyzed using AccuBrain® to generate the quantitative ratio of WMHs (WMHr, WMH volumes corrected by intracranial volume) and regional brain atrophy. We also divided WMHr into periventricular WMHr (PVWMHr) and deep WMHr (DWMHr) for the purpose of this study. The Cholinergic Pathways Hyperintensities Scale (CHIPS) scores were conducted by two evaluators. Independent t-test, Mann–Whitney U test, or χ2 test were used to compare the demographic characteristics, and Spearman correlation coefficient values were used to determine the association between WMHs and different regions of brain atrophy. Results Positive association between WMHr and quantitative medial temporal lobe atrophy (QMTA) (rs = 0.281, p = 0.011), temporal lobe atrophy (rs = 0.285, p = 0.011), and insular atrophy (rs = 0.406, p < 0.001) was found in the AD group before Bonferroni correction. PVWMHr contributed to these correlations. By separately analyzing the relationship between PVWMHr and brain atrophy, we found that there were still positive correlations after correction in QMTA (rs = 0.325, p = 0.003), temporal lobe atrophy (rs = 0.298, p = 0.007), and insular atrophy (rs = 0.429, p < 0.001) in AD group. Conclusion WMH severity tends to be associated with regional brain atrophy in patients with AD, especially with medial temporal lobe, temporal lobe, and insular lobe atrophy. PVWMHs were devoted to these correlations.
Collapse
Affiliation(s)
- Zhiyu Cao
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yingren Mai
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenli Fang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ming Lei
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yishan Luo
- BrainNow Research Institute, Shenzhen, China
| | - Lei Zhao
- BrainNow Research Institute, Shenzhen, China
| | - Wang Liao
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qun Yu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiaxin Xu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuting Ruan
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Songhua Xiao
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Vincent C. T. Mok
- BrainNow Research Institute, Shenzhen, China
- Division of Neurology, Department of Medicine and Therapeutics, Gerald Choa Neuroscience Centre, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Lin Shi
- BrainNow Research Institute, Shenzhen, China
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- *Correspondence: Lin Shi
| | - Jun Liu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Neurology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Jun Liu
| | | |
Collapse
|
16
|
Zhang J, Zhu Q, Wang X, Wang J. Effect of Donepezil on Vascular Dementia in Rats via PI3K/AKT Pathway. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background: Previous studies have shown that Donepezil has therapeutic effects on vascular dementia (VD). PI3K/AKT involves in oxidative stress injury and cell apoptosis. This study investigated whether Donepezil affects the neurological function and apoptosis of VD mice via
PI3K/AKT signaling. Methods: Mice were assigned into Sham group, VD group, VD+Donepezil groupfollowed by analysis of mice learning and memory ability by Water maze test, p-AKT expression by Western blot, Caspase-3 activity, MDA content, SOD activity and GSH-Px in hippocampus. HT22 cells
were cultured and separated into control group, I-R group and I-R+Donepezil group followed by measuring p-AKT level, ROS content and apoptosis. Results: Learning and memory abilities of VD group mice were significantly decreased, Caspase-3 activity and MDA in brain tissue were significantly
increased, along with decreased SOD activity, GSH-Px and p-AKT level. Donepezil treatment can significantly improve VD mice learning and memory ability, reduce Caspase-3 activity and MDA in brain tissue, increase SOD activity, GSH-Px and p-AKT level. In vitro, I-R treatment significantly
induced apoptosis of HT22 cells, increased ROS production and decreased p-AKT level. Donepezil treatment could up-regulate p-AKT in HT22 cells and reduce apoptosis and ROS production in HT22 cells. Conclusion: Donepezil improves the function of brain nerve in VD mice through regulating
PI3K/AKT pathway, thus reducing oxidative stress injury and apoptosis of brain nerve cells.
Collapse
Affiliation(s)
- Jianmin Zhang
- Department of Neurology, The First People’s Hospital of Fuyang District, Hangzhou, 311400, Zhejiang, China
| | - Qianwen Zhu
- Department of Neurology, The First People’s Hospital of Fuyang District, Hangzhou, 311400, Zhejiang, China
| | - Xingnan Wang
- Department of Neurology, The First People’s Hospital of Fuyang District, Hangzhou, 311400, Zhejiang, China
| | - Jian Wang
- Department of Neurology, The First People’s Hospital of Fuyang District, Hangzhou, 311400, Zhejiang, China
| |
Collapse
|
17
|
Study Design and Baseline Results in a Cohort Study to Identify Predictors for the Clinical Progression to Mild Cognitive Impairment or Dementia From Subjective Cognitive Decline (CoSCo) Study. Dement Neurocogn Disord 2022; 21:147-161. [PMID: 36407288 PMCID: PMC9644060 DOI: 10.12779/dnd.2022.21.4.147] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/24/2022] [Accepted: 10/28/2022] [Indexed: 11/09/2022] Open
Abstract
Background and Purpose Subjective cognitive decline (SCD) refers to the self-perception of cognitive decline with normal performance on objective neuropsychological tests. SCD, which is the first help-seeking stage and the last stage before the clinical disease stage, can be considered to be the most appropriate time for prevention and treatment. This study aimed to compare characteristics between the amyloid positive and amyloid negative groups of SCD patients. Methods A cohort study to identify predictors for the clinical progression to mild cognitive impairment (MCI) or dementia from subjective cognitive decline (CoSCo) study is a multicenter, prospective observational study conducted in the Republic of Korea. In total, 120 people aged 60 years or above who presented with a complaint of persistent cognitive decline were selected, and various risk factors were measured among these participants. Continuous variables were analyzed using the Wilcoxon rank-sum test, and categorical variables were analyzed using the χ2 test or Fisher’s exact test. Logistic regression models were used to assess the predictors of amyloid positivity. Results The multivariate logistic regression model indicated that amyloid positivity on PET was related to a lack of hypertension, atrophy of the left temporal lateral and entorhinal cortex, low body mass index, low waist circumference, less body and visceral fat, fast gait speed, and the presence of the apolipoprotein E ε4 allele in amnestic SCD patients. Conclusions The CoSCo study is still in progress, and the authors aim to identify the risk factors that are related to the progression of MCI or dementia in amnestic SCD patients through a two-year follow-up longitudinal study.
Collapse
|
18
|
Thorp EB, Flanagan ME, Popko B, DeBerge M. Resolving inflammatory links between myocardial infarction and vascular dementia. Semin Immunol 2022; 59:101600. [PMID: 35227567 PMCID: PMC10234261 DOI: 10.1016/j.smim.2022.101600] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 02/07/2022] [Accepted: 02/15/2022] [Indexed: 01/15/2023]
Abstract
Myocardial infarction is associated with increased risk for vascular dementia. In both myocardial infarction and vascular dementia, there is evidence that elevated inflammatory biomarkers are associated with worsened clinical outcomes. Myocardial infarction leads to a systemic inflammatory response, which may contribute to recruitment or activation of myeloid cells, including monocytes, microglia, and perivascular macrophages, within the central nervous system. However, our understanding of the causative roles for these cells linking cardiac injury to the development and progression of dementia is incomplete. Herein, we provide an overview of inflammatory cellular and molecular links between myocardial infarction and vascular dementia and discuss strategies to resolve inflammation after myocardial infarction to limit neurovascular injury.
Collapse
Affiliation(s)
- Edward B Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States.
| | - Margaret E Flanagan
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States; Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Brian Popko
- Department of Neurology, Division of Multiple Sclerosis and Neuroimmunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Matthew DeBerge
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States.
| |
Collapse
|
19
|
Du XL, Song L, Schulz PE, Xu H, Chan W. Risk of Developing Alzheimer's Disease and Related Dementias in Association with Cardiovascular Disease, Stroke, Hypertension, and Diabetes in a Large Cohort of Women with Breast Cancer and with up to 26 Years of Follow-Up. J Alzheimers Dis 2022; 87:415-432. [PMID: 35311707 PMCID: PMC9117151 DOI: 10.3233/jad-215657] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND No study on the long-term incidence of Alzheimer's disease (AD) and related dementias (ADRD) has been reported in women with breast cancer by vascular diseases. OBJECTIVE To determine the risk of ADRD in association with cardiovascular diseases (CVD), stroke, hypertension, and diabetes in women with breast cancer. METHODS Study identified 246,686 women diagnosed with breast cancer at age≥65 years in 1991-2015 from the Surveillance, Epidemiology, and End Results (SEER)-Medicare linked database. Women were free of ADRD at the time of cancer diagnosis and followed from 1991 to 2016. RESULTS Cumulative incidence of AD over 26 years of follow-up varied from 10.7% to 13.6% by CVD, stroke, hypertension, and diabetes. Cumulative incidence of ADRD was higher in those with CVD (40.75%) versus no-CVD (31.32%), stroke (40.24%) versus no-stroke (31.34%), hypertension (33.06%) versus no-hypertension (30.47%), and diabetes (33.38%) versus no-diabetes (31.77%). After adjusting for confounders, those with CVD (hazard ratio:1.30, 95% CI: 1.27-1.33), stroke (1.50,1.47-1.54), hypertension (1.08,1.06-1.09), and diabetes (1.26,1.24-1.29) had significantly higher risks of developing ADRD. Women aged 80-84, and≥85 had 5- and 7-fold higher risks of AD than those aged 65-69. As compared to white women, black women had a significantly higher risk of AD (1.21, 1.16-1.27), whereas Asians/Pacific-Islanders had a significantly lower risk of AD (0.77, 0.71-0.83). CONCLUSION In women with breast cancer, CVD, stroke, hypertension, and diabetes were associated with a significantly higher risk of developing any ADRD combined. The risk of ADRD was higher in black women and lower in Asian/Pacific-Islanders than white women.
Collapse
Affiliation(s)
- Xianglin L. Du
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Lulu Song
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Paul E. Schulz
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hua Xu
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Wenyaw Chan
- Department of Biostatistics and Data Science, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
20
|
Wang Y, Lou F, Li Y, Liu F, Wang Y, Cai L, Gordon ML, Zhang Y, Zhang N. Clinical, Neuropsychological, and Neuroimaging Characteristics of Amyloid- positive vs. Amyloid-negative Patients with Clinically Diagnosed Alzheimer's Disease and Amnestic Mild Cognitive Impairment. Curr Alzheimer Res 2021; 18:523-532. [PMID: 34598664 DOI: 10.2174/1567205018666211001113349] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/14/2021] [Accepted: 07/24/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND A significant proportion of patients with clinically diagnosed Alzheimer's Disease (AD) and an even higher proportion of patients with amnestic mild cognitive impairment (aMCI) do not show evidence of amyloid deposition on Positron Emission Tomography (PET) with amyloid-binding tracers such as 11C-labeled Pittsburgh Compound B (PiB). OBJECTIVE This study aimed to identify clinical, neuropsychological and neuroimaging factors that might suggest amyloid neuropathology in patients with clinically suspected AD or aMCI. METHODS Forty patients with mild to moderate AD and 23 patients with aMCI who were clinically diagnosed in our memory clinic and had PiB PET scans were included. Clinical, neuropsychological, and imaging characteristics, such as Medial Temporal lobe Atrophy (MTA) and White Matter Hyperintensities (WMH) on MRI and metabolic pattern on 18F-labeled fluorodeoxyglucose (FDG) PET, were compared between patients with PiB positive and negative PET results for AD, aMCI, and all subjects combined, respectively. RESULTS Compared with PiB positive patients, PiB negative patients had a higher prevalence of hypertension history, better performance on the Mini-Mental State Examination, the Rey Auditory Verbal Learning Test, and the Judgement of Line Orientation, lower score of MTA, and were less likely to have temporoparietal-predominant hypometabolism on FDG PET. Affective symptoms were less common in PiB negative patients diagnosed with AD, and the Animal Fluency Test score was higher in PiB negative patients diagnosed with aMCI. CONCLUSION In patients with clinically diagnosed AD or aMCI, absence of a history of hypertension, deficits in verbal learning and memory, visuospatial function, semantic verbal fluency, presence of affective symptoms, MTA on MRI, and temporoparietal hypometabolism on FDG PET suggested amyloid deposition in the brain.
Collapse
Affiliation(s)
- Yue Wang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, 154, Anshan Road, Tianjin, China
| | - Fanghua Lou
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, 154, Anshan Road, Tianjin, China
| | - Yonggang Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154, Anshan Road, Tianjin 300052, China
| | - Fang Liu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, 154, Anshan Road, Tianjin, China
| | - Ying Wang
- PET/CT Center, Tianjin Medical University General Hospital, 154, Anshan Road, Tianjin, China
| | - Li Cai
- PET/CT Center, Tianjin Medical University General Hospital, 154, Anshan Road, Tianjin, China
| | - Marc L Gordon
- The Litwin-Zucker Research Center, The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY, United States
| | - Yuanyuan Zhang
- Department of Radiology, Tianjin Medical University General Hospital, 154, Anshan Road, Tianjin, China
| | - Nan Zhang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, 154, Anshan Road, Tianjin, China
| |
Collapse
|
21
|
Komici K, Femminella GD, Bencivenga L, Rengo G, Pagano G. Diabetes Mellitus and Parkinson's Disease: A Systematic Review and Meta-Analyses. JOURNAL OF PARKINSONS DISEASE 2021; 11:1585-1596. [PMID: 34486987 DOI: 10.3233/jpd-212725] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND A link between diabetes mellitus (DM) and Parkinson's disease (PD) have been proposed but evidence are sparse and inconsistent. OBJECTIVE Perform a systematic review of all evidence that link DM and PD characterising the prevalence of DM in PD patients, the risk of developing PD in DM patients and the influence of DM on PD severity and progression. METHODS MEDLINE, Scopus, and Cochrane Library from inception to June 30, 2021 were searched. Studies reporting prevalence, incidence, severity and disease progression of DM and PD were included. Prevalence of DM in PD and incidence of PD in DM patients, and characteristics of PD. RESULTS A total of 21 studies (n = 11,396) included data on DM prevalence in PD patients, 12 studies (n = 17,797,221) included data on incidence of PD in DM patients, and 10 studies (n = 2,482) included data on DM impact on PD severity and disease progression. The prevalence of DM in PD patients was 10.02 %, (95%C.I. 7.88 -12.16), DM patients showed a higher risk of developing PD (OR: 1.34 95%CI 1.26-1.43 p < 0.0001) compared to non-DM, and PD patients with DM showed a greater severity of motor symptoms, with higher Hoehn and Yahr stage (SMD: 0.36 95%CI 0.12-0.60; p < 0.001) and higher UPDRS (SMD 0.60 95%CI 0.28-0.92; p < 0.001) compared with PD patients without DM. CONCLUSION Although the prevalence of DM in PD patients is similar to the general population, patients with DM have a higher risk of developing PD, and the presence of DM is associated with greater PD severity and faster progression, which suggests that DM may be a facilitating factor of neurodegeneration.
Collapse
Affiliation(s)
- Klara Komici
- Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | | | - Leonardo Bencivenga
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy.,Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy.,Istituti Clinici Scientifici Maugeri SpA Società Benefit (ICS Maugeri SpA SB), Scientific Institute of Telese Terme, Telese Terme (BN), Italy
| | - Gennaro Pagano
- King's College London, London, UK.,Roche Pharma Research and Early Development (pRED), Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center, Basel, Switzerland
| |
Collapse
|
22
|
Graff BJ, Payne SJ, El-Bouri WK. The Ageing Brain: Investigating the Role of Age in Changes to the Human Cerebral Microvasculature With an in silico Model. Front Aging Neurosci 2021; 13:632521. [PMID: 34421568 PMCID: PMC8374868 DOI: 10.3389/fnagi.2021.632521] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 07/12/2021] [Indexed: 11/25/2022] Open
Abstract
Ageing causes extensive structural changes to the human cerebral microvasculature, which have a significant effect on capillary bed perfusion and oxygen transport. Current models of brain capillary networks in the literature focus on healthy adult brains and do not capture the effects of ageing, which is critical when studying neurodegenerative diseases. This study builds upon a statistically accurate model of the human cerebral microvasculature based on ex-vivo morphological data. This model is adapted for “healthy” ageing using in-vivo measurements from mice at three distinct age groups—young, middle-aged, and old. From this new model, blood and molecular exchange parameters are calculated such as permeability and surface-area-to-volume ratio, and compared across the three age groups. The ability to alter the model vessel-by-vessel is used to create a continuous gradient of ageing. It was found that surface-area-to-volume ratio reduced in old age by 6% and permeability by 24% from middle-age to old age, and variability within the networks also increased with age. The ageing gradient indicated a threshold in the ageing process around 75 years old, after which small changes have an amplified effect on blood flow properties. This gradient enables comparison of studies measuring cerebral properties at discrete points in time. The response of middle aged and old aged capillary beds to micro-emboli showed a lower robustness of the old age capillary bed to vessel occlusion. As the brain ages, there is thus increased vulnerability of the microvasculature—with a “tipping point” beyond which further remodeling of the microvasculature has exaggerated effects on the brain. When developing in-silico models of the brain, age is a very important consideration to accurately assess risk factors for cognitive decline and isolate early biomarkers of microvascular health.
Collapse
Affiliation(s)
- Barnaby J Graff
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Stephen J Payne
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| | - Wahbi K El-Bouri
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom.,Liverpool Centre for Cardiovascular Science, University of Liverpool & Liverpool Heart and Chest Hospital, Liverpool, United Kingdom.,Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
23
|
Niu F, Sharma A, Wang Z, Feng L, Muresanu DF, Sahib S, Tian ZR, Lafuente JV, Buzoianu AD, Castellani RJ, Nozari A, Menon PK, Patnaik R, Wiklund L, Sharma HS. Nanodelivery of oxiracetam enhances memory, functional recovery and induces neuroprotection following concussive head injury. PROGRESS IN BRAIN RESEARCH 2021; 265:139-230. [PMID: 34560921 DOI: 10.1016/bs.pbr.2021.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Military personnel are the most susceptible to concussive head injury (CHI) caused by explosion, blast or missile or blunt head trauma. Mild to moderate CHI could induce lifetime functional and cognitive disturbances causing significant decrease in quality of life. Severe CHI leads to instant death and lifetime paralysis. Thus, further exploration of novel therapeutic agents or new features of known pharmacological agents are needed to enhance quality of life of CHI victims. Previous reports from our laboratory showed that mild CHI induced by weight drop technique causing an impact of 0.224N results in profound progressive functional deficit, memory impairment and brain pathology from 5h after trauma that continued over several weeks of injury. In this investigation we report that TiO2 nanowired delivery of oxiracetam (50mg/kg, i.p.) daily for 5 days after CHI resulted in significant improvement of functional deficit on the 8th day. This was observed using Rota Rod treadmill, memory improvement assessed by the time spent in finding hidden platform under water. The motor function improvement is seen in oxiracetam treated CHI group by placing forepaw on an inclined mesh walking and foot print analysis for stride length and distance between hind feet. TiO2-nanowired oxiracetam also induced marked improvements in the cerebral blood flow, reduction in the BBB breakdown and edema formation as well as neuroprotection of neuronal, glial and myelin damages caused by CHI at light and electron microscopy on the 7th day after 5 days TiO2 oxiracetam treatment. Adverse biochemical events such as upregulation of CSF nitrite and nitrate, IL-6, TNF-a and p-Tau are also reduced significantly in oxiracetam treated CHI group. On the other hand post treatment of 100mg/kg dose of normal oxiracetam in identical conditions after CHI is needed to show slight but significant neuroprotection together with mild recovery of memory function and functional deficits on the 8th day. These observations are the first to point out that nanowired delivery of oxiracetam has superior neuroprotective ability in CHI. These results indicate a promising clinical future of TiO2 oxiracetam in treating CHI patients for better quality of life and neurorehabilitation, not reported earlier.
Collapse
Affiliation(s)
- Feng Niu
- CSPC NBP Pharmaceutical Medicine, Shijiazhuang, China
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Zhenguo Wang
- CSPC NBP Pharmaceutical Medicine, Shijiazhuang, China
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - Preeti K Menon
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
24
|
Thomassen JQ, Tolstrup JS, Nordestgaard BG, Tybjærg-Hansen A, Frikke-Schmidt R. Plasma Concentrations of Magnesium and Risk of Dementia: A General Population Study of 102 648 Individuals. Clin Chem 2021; 67:899-911. [PMID: 33846733 DOI: 10.1093/clinchem/hvab041] [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] [Received: 12/14/2020] [Accepted: 02/18/2021] [Indexed: 01/13/2023]
Abstract
BACKGROUND Low and high concentrations of plasma magnesium are associated with increased risk of future all-cause dementia; however, the underlying reasons remain elusive. The magnesium ion is an important electrolyte serving as a cofactor in many enzymatic processes in the human organism. Magnesium affects both neuronal and vascular functions. We investigated the associations of plasma concentrations of magnesium associate with common subtypes of dementia as Alzheimer dementia and non-Alzheimer dementia, and potential pathways by which magnesium may affect risk of dementia. METHODS Plasma concentrations of magnesium were measured in 102 648 individuals from the Copenhagen General Population Study. Cox regression and natural effects mediation analyses evaluated associations with either Alzheimer dementia or non-Alzheimer dementia. RESULTS Multifactorially adjusted hazard ratios for non-Alzheimer dementia were 1.50(95% confidence interval (CI):1.21-1.87) for the lowest and 1.34(1.07-1.69) for the highest vs the fourth quintile (reference) of plasma magnesium concentrations. Diabetes, cumulated smoking, stroke, and systolic blood pressure mediated 10.4%(3.1-22.8%), 6.8%(1.2-14.0%), 1.3%(0.1-3.6%), and 1.0%(0.2-2.6%), respectively, in the lowest quintile, whereas stroke mediated 3.2%(0.4-11.9%) in the highest quintile. No associations were observed for Alzheimer dementia. CONCLUSIONS Low and high plasma magnesium concentrations were associated with high risk of vascular-related non-Alzheimer dementia, with the lowest risk observed at a concentration of 2.07 mg/dL (0.85 mmol/L). No association was observed for Alzheimer dementia. Mediation analysis suggested that diabetes may be in the causal pathway between low plasma magnesium concentrations and high risk of non-Alzheimer dementia, while cumulated smoking, stroke, and systolic blood pressure played minor mediating roles.
Collapse
Affiliation(s)
| | - Janne S Tolstrup
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Herlev, Denmark.,The Copenhagen General Population Study, Herlev and Gentofte Hospital, Herlev, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Tybjærg-Hansen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark.,The Copenhagen General Population Study, Herlev and Gentofte Hospital, Herlev, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark.,The Copenhagen General Population Study, Herlev and Gentofte Hospital, Herlev, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW The current review evaluates the recent literature on the impact of metabolic dysfunction in human cognition, focusing on epidemiological studies and meta-analyses of these. RECENT FINDINGS Worldwide around 50 million people live with dementia, a number projected to triple by 2050. Recent reports from the Lancet Commission suggest that 40% of dementia cases may be preventable primarily by focusing on well established metabolic dysfunction components and cardiovascular risk factors. SUMMARY There is robust evidence that type 2 diabetes and midlife hypertension increase risk of dementia in late life. Obesity and elevated levels of LDL cholesterol in midlife probably increase risk of dementia, but further research is needed in these areas. Physical activity, diet, alcohol, and smoking might also influence the risk of dementia through their effect on metabolic dysfunction. A key recommendation is to be ambitious about prevention, focusing on interventions to promote healthier lifestyles combating metabolic dysfunction. Only comprehensive multidomain and staff-requiring interventions are however efficient to maintain or improve cognition in at-risk individuals and will be unrealistic economic burdens for most societies to implement. Therefore, a risk score that identifies high-risk individuals will enable a targeted early intensive intervention toward those high-risk individuals that will benefit the most from a prevention against cardiovascular risk factors and metabolic dysfunction.
Collapse
Affiliation(s)
| | | | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Rigshospitalet
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
26
|
Nguyen DH, Cunningham JT, Sumien N. Estrogen receptor involvement in vascular cognitive impairment and vascular dementia pathogenesis and treatment. GeroScience 2021; 43:159-166. [PMID: 32902819 PMCID: PMC8050128 DOI: 10.1007/s11357-020-00263-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/02/2020] [Indexed: 01/26/2023] Open
Abstract
Vascular cognitive impairment (VCI) is a term that encompasses a continuum of cognitive disorders with cerebrovascular pathology contribution, ranging from mild cognitive impairment to vascular dementia (VaD). VCI and VaD, thus, represent an interesting intersection between cardiovascular disease and neurodegenerative disorders such as Alzheimer's disease (AD) and a rising area of research in recent years. Although VCI and VaD research has identified various causes and explanations for disease development, many aspects remain unclear, particularly sex differences in VCI (e.g., epidemiology), unlike those available for cardiovascular disease and AD. Despite limited information in the literature, several studies have observed an association of estrogen receptor (ER) polymorphisms and VaD. If further explored, this association could provide valuable insights for novel therapeutic approaches. This review aims to provide a brief epidemiological overview and subsequent discussion exploring concepts of brain aging and involvement of estrogen receptors in potential mechanisms of VCI/VaD pathogenesis and treatment development.
Collapse
Affiliation(s)
- Dianna H Nguyen
- Department of Physiology and Anatomy, UNT Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107, USA
- Texas College of Osteopathic Medicine, UNT Health Science Center, Fort Worth, TX, USA
| | - J Thomas Cunningham
- Department of Physiology and Anatomy, UNT Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX, 76107, USA.
| | - Nathalie Sumien
- Department of Pharmacology and Neuroscience, UNT Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
27
|
Yang D, Masurkar AV. Clinical Profiles of Arteriolosclerosis and Alzheimer Disease at Mild Cognitive Impairment and Mild Dementia in a National Neuropathology Cohort. Alzheimer Dis Assoc Disord 2021; 35:14-22. [PMID: 32925200 PMCID: PMC7904566 DOI: 10.1097/wad.0000000000000411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 08/21/2020] [Indexed: 11/26/2022]
Abstract
OBJECTIVE We sought to evaluate early clinical differences between cerebral arteriolosclerosis (pARTE), Alzheimer disease (pAD), and AD with arteriolosclerosis (ADARTE). METHODS Using National Alzheimer's Coordinating Center neuropathology diagnoses, we defined pARTE (n=21), pAD (n=203), and ADARTE (n=158) groups. We compared demographics, medical history, psychometrics, neuropsychiatric symptoms, and apolipoprotein E (APOE) allele variants across neuropathology groups. Retrospective timepoints were first evaluation with Global Clinical Dementia Rating (CDR) score of 0.5 and 1.0, via the CDR Dementia Staging Instrument, corresponding to mild cognitive impairment (MCI) and mild dementia, respectively. RESULTS In MCI, clinical differences were minimal but pARTE subjects were older, had later onset cognitive decline, and progressed less severely than pAD. In mild dementia, pAD subjects were younger and had earlier onset of decline. Neuropsychiatric (depression) and psychometric (Logical Memory Delayed Recall, Trails B) differences also emerged between the groups. In MCI, APOE4 associated with worse Logical Memory Delayed Recall in pAD and ADARTE. In mild dementia, APOE4 associated with better animal fluency in pAD, but with better Trails A performance and more neuropsychiatric symptoms (Neuropsychiatric Inventory Questionnaire) in ADARTE. CONCLUSIONS Differences between pARTE, pAD, and ADARTE emerge at mild dementia rather than MCI. APOE4 has varied cognitive and psychiatric impact dependent on neuropathology group and stage.
Collapse
Affiliation(s)
| | - Arjun V Masurkar
- Department of Neurology, New York University School of Medicine, Center for Cognitive Neurology, New York, NY
| |
Collapse
|
28
|
Ek Olofsson H, Haglund M, Englund E. Are cortical microvascular raspberries caused by cerebral hypoperfusion? An exploratory pathological study. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2021; 2:100026. [PMID: 36324730 PMCID: PMC9616238 DOI: 10.1016/j.cccb.2021.100026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/03/2021] [Accepted: 08/15/2021] [Indexed: 12/03/2022]
Abstract
An exploratory study on the ‘raspberry’, a type of cortical microvascular formation. Hypothesis: raspberries form by angiogenesis induced by cerebral hypoperfusion. Are raspberries associated with clinical or pathological markers of hypoperfusion? Data: histopathological raspberry quantification, medical records, autopsy reports. Raspberries were associated with atherosclerosis of the basal cerebral arteries.
Introduction This retrospective study investigated a cortical microvascular formation, termed a ‘raspberry’ due to its appearance under a bright-field microscope. We examined whether there is support for the hypothesis that raspberry formation is an angiogenic process induced by cerebral hypoperfusion. Materials and Methods Raspberries were manually quantified in haematoxylin and eosin-stained cortical sections from the anterior frontal lobe of deceased individuals who had undergone a diagnostic neuropathological examination at the Department of Pathology, Lund, Sweden, during April 2019–January 2021. Subjects represented consecutively received cases during this 22-month period. The raspberry density was compared between subjects according to variables collected from medical records and autopsy reports: age, sex, hypertension, diabetes mellitus, atrial fibrillation, orthostatic hypotension, chronic heart failure, acute circulatory failure, aortic atherosclerosis, atherosclerosis of the basal cerebral arteries (referred to as ‘cerebral atherosclerosis’), cerebral small vessel disease, cerebral amyloid angiopathy, cerebral infarction, and ischaemic white matter disease. Results 62 subjects were included. The mean age was 71.9 years (range 46–97 years). 21 subjects (33.9%) were female. Independent-samples t-test showed a higher raspberry density in subjects with cerebral atherosclerosis (p = 0.029; 95% CI 0.7, 11.6 raspberries/cm²). The higher raspberry density in subjects with cerebral atherosclerosis remained in multiple linear regression (p = 0.003; 95% CI 2.3, 11.1 raspberries/cm²). Conclusion This exploratory study indicates that cortical raspberries could be associated with cerebral atherosclerosis. The remaining results were inconclusive but motivate further examination of variables such as acute circulatory failure.
Collapse
|
29
|
Nyúl-Tóth Á, Tarantini S, Kiss T, Toth P, Galvan V, Tarantini A, Yabluchanskiy A, Csiszar A, Ungvari Z. Increases in hypertension-induced cerebral microhemorrhages exacerbate gait dysfunction in a mouse model of Alzheimer's disease. GeroScience 2020; 42:1685-1698. [PMID: 32844283 PMCID: PMC7732885 DOI: 10.1007/s11357-020-00256-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/17/2020] [Indexed: 12/17/2022] Open
Abstract
Clinical studies show that cerebral amyloid angiopathy (CAA) associated with Alzheimer's disease (AD) and arterial hypertension are independent risk factors for cerebral microhemorrhages (CMHs). To test the hypothesis that amyloid pathology and hypertension interact to promote the development of CMHs, we induced hypertension in the Tg2576 mouse model of AD and respective controls by treatment with angiotensin II (Ang II) and the NO synthesis inhibitor L-NAME. The number, size, localization, and neurological consequences (gait alterations) of CMHs were compared. We found that compared to control mice, in TG2576 mice, the same level of hypertension led to significantly increased CMH burden and exacerbation of CMH-related gait alterations. In hypertensive TG2576 mice, CMHs were predominantly located in the cerebral cortex at the cortical-subcortical boundary, mimicking the clinical picture seen in patients with CAA. Collectively, amyloid pathologies exacerbate the effects of hypertension, promoting the genesis of CMHs, which likely contribute to their deleterious effects on cognitive function. Therapeutic strategies for prevention of CMHs that reduce blood pressure and preserve microvascular integrity are expected to exert neuroprotective effects in high-risk elderly AD patients.
Collapse
Affiliation(s)
- Ádám Nyúl-Tóth
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Tamas Kiss
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Theoretical Medicine Doctoral School/Departments of Medical Physics and Informatics & Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary
| | - Peter Toth
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Doctoral School of Clinical Medicine, Department of Neurosurgery and Szentagothai Research Center, Medical School, University of Pecs, Pecs, Hungary
| | - Veronica Galvan
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- South Texas Veterans Health Care System, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Amber Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Theoretical Medicine Doctoral School/Departments of Medical Physics and Informatics & Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary.
- International Training Program in Geroscience, Theoretical Medicine Doctoral School/Departments of Medical Physics and Informatics & Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary.
- Department of Biochemistry and Molecular Biology, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
30
|
Javanshiri K, Haglund M, Englund E. Cardiovascular Disease, Diabetes Mellitus, and Hypertension in Lewy Body Disease: A Comparison with Other Dementia Disorders. J Alzheimers Dis 2020; 71:851-859. [PMID: 31450500 PMCID: PMC6839595 DOI: 10.3233/jad-190485] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background: Research concerning the potential roles of cardiovascular disease (CaVD) and diabetes mellitus (DM) as risk factors for Lewy body disease (LBD) is limited. These disorders are, however, established risk factors for vascular dementia (VaD) and have been proposed as risk factors for Alzheimer’s disease (AD). Objective: The aim of this study was to investigate the prevalence of CaVD and DM in LBD and compare the results with previous findings in cases with AD, VaD, and mixed AD-VaD (MD). Methods: Autopsy reports at the Clinical Department of Pathology in Lund from 2001–2018 were analyzed. All cases with a complete neuropathological diagnosis of LBD were selected, not distinguishing between subjects with clinical Parkinson disease dementia and dementia with Lewy bodies, on the condition of a clinical diagnosis of dementia. Clinical data were retrieved through the patients’ medical records and the Swedish National Diabetes Register (NDR) and compared with those of the AD, VaD, and MD cases. Results: In LBD, there was less CaVD, significantly less DM (p = 0.002) and likewise significantly less hypertension (p < 0.001) than in VaD. The results of the LBD group were consistent with the results of the AD group. Conclusion: Our findings of a low prevalence of CaVD and CaVD risk factors in LBD and in AD argue against the association between these risk factors and their contribution to the development of neurodegenerative diseases.
Collapse
Affiliation(s)
- Keivan Javanshiri
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Sweden; Department of Clinical Pathology, Lab Medicine, Region Skane, Sweden
| | - Mattias Haglund
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Sweden; Department of Clinical Pathology, Lab Medicine, Region Skane, Sweden
| | - Elisabet Englund
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Sweden; Department of Clinical Pathology, Lab Medicine, Region Skane, Sweden
| |
Collapse
|
31
|
Jørgensen IF, Aguayo‐Orozco A, Lademann M, Brunak S. Age-stratified longitudinal study of Alzheimer's and vascular dementia patients. Alzheimers Dement 2020; 16:908-917. [PMID: 32342671 PMCID: PMC7383608 DOI: 10.1002/alz.12091] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/17/2019] [Accepted: 02/21/2020] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Similar symptoms, comorbidities and suboptimal diagnostic tests make the distinction between different types of dementia difficult, although this is essential for improved work-up and treatment optimization. METHODS We calculated temporal disease trajectories of earlier multi-morbidities in Alzheimer's disease (AD) dementia and vascular dementia (VaD) patients using the Danish National Patient Registry covering all hospital encounters in Denmark (1994 to 2016). Subsequently, we reduced the comorbidity space dimensionality using a non-linear technique, uniform manifold approximation and projection. RESULTS We found 49,112 and 24,101 patients that were diagnosed with AD or VaD, respectively. Temporal disease trajectories showed very similar disease patterns before the dementia diagnosis. Stratifying patients by age and reducing the comorbidity space to two dimensions, showed better discrimination between AD and VaD patients in early-onset dementia. DISCUSSION Similar age-associated comorbidities, the phenomenon of mixed dementia, and misdiagnosis create great challenges in discriminating between classical subtypes of dementia.
Collapse
Affiliation(s)
- Isabella Friis Jørgensen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical SciencesUniversity of CopenhagenBlegdamsvej 3BCopenhagenDenmark
| | - Alejandro Aguayo‐Orozco
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical SciencesUniversity of CopenhagenBlegdamsvej 3BCopenhagenDenmark
| | - Mette Lademann
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical SciencesUniversity of CopenhagenBlegdamsvej 3BCopenhagenDenmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical SciencesUniversity of CopenhagenBlegdamsvej 3BCopenhagenDenmark
| |
Collapse
|
32
|
Dynamic Changes of Beclin-1 in the Hippocampus of Male Mice with Vascular Dementia at Different Time Points. J Mol Neurosci 2020; 70:1611-1618. [DOI: 10.1007/s12031-020-01591-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 05/13/2020] [Indexed: 12/14/2022]
|
33
|
Thomassen JQ, Tolstrup JS, Benn M, Frikke-Schmidt R. Type-2 diabetes and risk of dementia: observational and Mendelian randomisation studies in 1 million individuals. Epidemiol Psychiatr Sci 2020; 29:e118. [PMID: 32326995 PMCID: PMC7214711 DOI: 10.1017/s2045796020000347] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 02/21/2020] [Accepted: 03/22/2020] [Indexed: 01/10/2023] Open
Abstract
AIMS In observational studies, type-2 diabetes is associated with increased risk of dementia; however, the causal nature of this association remains unanswered. In an unselected nationwide study of all Danes, we wanted to test whether type-2 diabetes is associated with dementia subtypes, and to test whether potential associations are of a causal nature. METHODS In the current study of nationwide observational registry data in all Danes above the age of 65 years (n = 784 434) combined with genetic consortia data on 213 370 individuals, we investigated the associations between type-2 diabetes and Alzheimer's disease, vascular dementia, unspecified dementia and all-cause dementia, and whether observational associations were of a causal nature by applying a two-sample Mendelian randomisation strategy. We addressed key biases inherent in Mendelian randomisation approaches. RESULTS Important confounders (age, ethnicity, size of community, region, civil status and education level) were captured on all 784 434 individuals and adjusted for in the models. Multifactorial adjusted hazard ratios were 1.13 (1.06-1.21) for Alzheimer's disease, 1.98 (1.83-2.14) for vascular dementia, 1.53 (1.48-1.59) for unspecified dementia and 1.48 (1.44-1.53) for all-cause dementia in persons with type-2 diabetes v. without. Results were similar for men and women. The two-sample Mendelian randomisation estimate for the association between the genetic instrument and Alzheimer's disease was 1.04 (0.98-1.10), consistent with sensitivity estimates, addressing pleiotropy, measurement bias and weak instrument bias. CONCLUSIONS In a nationwide study of all Danes above the age of 65 years, we show that type-2 diabetes is associated with major subtypes of dementia - with particularly strong associations for vascular dementia and unspecified dementia - the two types of dementia with the most obvious vascular pathologies. Although the present two-sample Mendelian randomisation approach using genetic consortia data suggests that type-2 diabetes is not a direct cause of Alzheimer's disease, we were unable to test the causal nature of type-2 diabetes for vascular dementia and unspecified dementia, because no publicly available genetic consortia data yet exist for these dementia endpoints. The causal nature of type-2 diabetes for dementia with vascular pathologies is pivotal questions to solve for future public health recommendations and therapeutic advice.
Collapse
Affiliation(s)
| | | | - Marianne Benn
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
34
|
Garrett MD. Multiple Causes of Dementia as Engineered Senescence. EUROPEAN JOURNAL OF MEDICAL AND HEALTH SCIENCES 2020; 2. [DOI: 10.24018/ejmed.2020.2.2.227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
All traumas—cranial, cardiovascular, hormone, viral, bacterial, fungi, parasites, misfolded protein, genetic, behavior, environmental and medication—affect the brain. This paper itemizes studies showing the many different causes of dementia including Alzheimer’s disease. Causes interact with each other, act sequentially by preparing the optimal conditions for its successor, initiate other diseases, allow for other traumas to accumulate and degrade protective features of the brain. Since such age-related cognitive impairment is not exclusively a human attribute there might be support for an evolutionary theory of dementia. Relying on theories of antagonistic pleiotropy and polymorphism, the brain has been designed to sequester trauma. Because of increased longevity, the short-term tactic of sequestering trauma becomes a long-term liability. We are engineered to sequester these insults until a tipping point is reached. Dementia is an evolutionary trade-off for longevity. We cannot cure dementia without understanding the overall biology of aging.
Collapse
|
35
|
Santiago JA, Bottero V, Potashkin JA. Transcriptomic and Network Analysis Highlight the Association of Diabetes at Different Stages of Alzheimer's Disease. Front Neurosci 2019; 13:1273. [PMID: 31849586 PMCID: PMC6895844 DOI: 10.3389/fnins.2019.01273] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/11/2019] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) and type 2 diabetes (T2D) are among the most prevalent chronic diseases affecting the aging population. Extensive research evidence indicates that T2D is a well-established risk factor for AD; however, the molecular mechanisms underlying this association have not been fully elucidated. Furthermore, how T2D may contribute to the progression of AD is a subject of extensive investigation. In this study, we compared the blood transcriptome of patients with mild cognitive impairment (MCI), AD, and advanced AD to those afflicted with T2D to unveil shared and unique pathways and potential therapeutic targets. Blood transcriptomic analyses revealed a positive correlation between gene expression profiles of MCI, AD, and T2D in seven independent microarrays. Interestingly, gene expression profiles from women with advanced AD correlated negatively with T2D, suggesting sex-specific differences in T2D as a risk factor for AD. Network and pathway analysis revealed that shared molecular networks between MCI and T2D were predominantly enriched in inflammation and infectious diseases whereas those networks shared between overt AD and T2D were involved in the phosphatidylinositol 3-kinase and protein kinase B/Akt (PI3K-AKT) signaling pathway, a major mediator of insulin signaling in the body. The PI3K-AKT signaling pathway became more significantly dysregulated in the advanced AD and T2D shared network. Furthermore, endocrine resistance and atherosclerosis pathways emerged as dysregulated pathways in the advanced AD and T2D shared network. Interestingly, network analysis of shared differentially expressed genes between children with T2D and MCI subjects identified forkhead box O3 (FOXO3) as a central transcriptional regulator, suggesting that it may be a potential therapeutic target for early intervention in AD. Collectively, these results suggest that T2D may be implicated at different stages of AD through different molecular pathways disrupted during the preclinical phase of AD and more advanced stages of the disease.
Collapse
Affiliation(s)
| | - Virginie Bottero
- Department of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Judith A Potashkin
- Department of Cellular and Molecular Pharmacology, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| |
Collapse
|
36
|
Doney ASF, Bonney W, Jefferson E, Walesby KE, Bittern R, Trucco E, Connelly P, McCrimmon RJ, Palmer CNA. Investigating the Relationship Between Type 2 Diabetes and Dementia Using Electronic Medical Records in the GoDARTS Bioresource. Diabetes Care 2019; 42:1973-1980. [PMID: 31391202 DOI: 10.2337/dc19-0380] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 07/17/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To investigate the impact of type 2 diabetes on incidence of major dementia subtypes, Alzheimer and vascular dementia, using electronic medical records (EMR) in the GoDARTS bioresource. RESEARCH DESIGN AND METHODS GoDARTS (Genetics of Diabetes Audit and Research in Tayside Scotland) comprises a large case-control study of type 2 diabetes with longitudinal follow-up in EMR. Dementia case subjects after recruitment were passively identified in the EMR, and using a combination of case note review, an Alzheimer-specific weighted genetic risk score (wGRS), and APOE4 genotype, we validated major dementia subtypes. We undertook a retrospective matched cohort study to determine the risk of type 2 diabetes status for incident dementia accounting for competing risk of death. RESULTS Type 2 diabetes status was associated with a significant risk of any dementia (cause-specific hazard ratio [csHR] 1.46, 95% CI 1.31-1.64), which was attenuated, but still significant, when competing risk of death was accounted for (subdistribution [sd]HR 1.26, 95% CI 1.13-1.41). The accuracy of EMR-defined cases of Alzheimer or vascular dementia was high-positive predictive value (PPV) 86.4% and PPV 72.8%, respectively-and wGRS significantly predicted Alzheimer dementia (HR 1.23, 95% CI 1.12-1.34) but not vascular dementia (HR 1.02, 95% CI 0.91-1.15). Conversely, type 2 diabetes was strongly associated with vascular dementia (csHR 2.47, 95% C 1.92-3.18) but not Alzheimer dementia, particularly after competing risk of death was accounted for (sdHR 1.02, 95% CI 0.87-1.18). CONCLUSIONS Our study indicates that type 2 diabetes is associated with an increased risk of vascular dementia but not with an increased risk of Alzheimer dementia and highlights the potential value of bioresources linked to EMR to study dementia.
Collapse
Affiliation(s)
- Alex S F Doney
- Division of Population Health and Genomics, Ninewells Hospital & Medical School, University of Dundee, Dundee, Scotland, U.K.
| | - Wilfred Bonney
- Health Informatics Centre, Ninewells Hospital & Medical School, University of Dundee, Dundee, Scotland, U.K
| | - Emily Jefferson
- Health Informatics Centre, Ninewells Hospital & Medical School, University of Dundee, Dundee, Scotland, U.K
| | - Katherine E Walesby
- Alzheimer Scotland Dementia Research Centre and Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, Scotland, U.K
| | - Rachel Bittern
- Health Informatics Centre, Ninewells Hospital & Medical School, University of Dundee, Dundee, Scotland, U.K
| | - Emanuele Trucco
- VAMPIRE Project, Computing, School of Science and Engineering, University of Dundee, Scotland, U.K
| | - Peter Connelly
- NHS Tayside, Scotland, U.K.,Department of Neuroscience, University of Dundee, Dundee, Scotland, U.K.,Department of Applied Social Science, University of Stirling, Stirling, Scotland, U.K
| | - Rory J McCrimmon
- Division of Population Health and Genomics, Ninewells Hospital & Medical School, University of Dundee, Dundee, Scotland, U.K
| | - Colin N A Palmer
- Division of Population Health and Genomics, Ninewells Hospital & Medical School, University of Dundee, Dundee, Scotland, U.K
| |
Collapse
|
37
|
Duggal P, Mehan S. Neuroprotective Approach of Anti-Cancer Microtubule Stabilizers Against Tauopathy Associated Dementia: Current Status of Clinical and Preclinical Findings. J Alzheimers Dis Rep 2019; 3:179-218. [PMID: 31435618 PMCID: PMC6700530 DOI: 10.3233/adr-190125] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neuronal microtubule (MT) tau protein provides cytoskeleton to neuronal cells and plays a vital role including maintenance of cell shape, intracellular transport, and cell division. Tau hyperphosphorylation mediates MT destabilization resulting in axonopathy and neurotransmitter deficit, and ultimately causing Alzheimer’s disease (AD), a dementing disorder affecting vast geriatric populations worldwide, characterized by the existence of extracellular amyloid plaques and intracellular neurofibrillary tangles in a hyperphosphorylated state. Pre-clinically, streptozotocin stereotaxically mimics the behavioral and biochemical alterations similar to AD associated with tau pathology resulting in MT assembly defects, which proceed neuropathological cascades. Accessible interventions like cholinesterase inhibitors and NMDA antagonist clinically provides only symptomatic relief. Involvement of microtubule stabilizers (MTS) prevents tauopathy particularly by targeting MT oriented cytoskeleton and promotes polymerization of tubulin protein. Multiple in vitro and in vivo research studies have shown that MTS can hold substantial potential for the treatment of AD-related tauopathy dementias through restoration of tau function and axonal transport. Moreover, anti-cancer taxane derivatives and epothiolones may have potential to ameliorate MT destabilization and prevent the neuronal structural and functional alterations associated with tauopathies. Therefore, this current review strictly focuses on exploration of various clinical and pre-clinical features available for AD to understand the neuropathological mechanisms as well as introduce pharmacological interventions associated with MT stabilization. MTS from diverse natural sources continue to be of value in the treatment of cancer, suggesting that these agents have potential to be of interest in the treatment of AD-related tauopathy dementia in the future.
Collapse
Affiliation(s)
- Pallavi Duggal
- Neuropharmacology Division, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
38
|
Ghannam M, Oral H. Atrial fibrillation and dementia: Understanding the risks in hopes of brighter tomorrows. Heart Rhythm 2019; 16:10-11. [DOI: 10.1016/j.hrthm.2018.10.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Indexed: 10/27/2022]
|