1
|
Smith EE, Biessels GJ, Gao V, Gottesman RF, Liesz A, Parikh NS, Iadecola C. Systemic determinants of brain health in ageing. Nat Rev Neurol 2024; 20:647-659. [PMID: 39375564 DOI: 10.1038/s41582-024-01016-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2024] [Indexed: 10/09/2024]
Abstract
Preservation of brain health is a worldwide priority. The traditional view is that the major threats to the ageing brain lie within the brain itself. Consequently, therapeutic approaches have focused on protecting the brain from these presumably intrinsic pathogenic processes. However, an increasing body of evidence has unveiled a previously under-recognized contribution of peripheral organs to brain dysfunction and damage. Thus, in addition to the well-known impact of diseases of the heart and endocrine glands on the brain, accumulating data suggest that dysfunction of other organs, such as gut, liver, kidney and lung, substantially affects the development and clinical manifestation of age-related brain pathologies. In this Review, a framework is provided to indicate how organ dysfunction can alter brain homeostasis and promote neurodegeneration, with a focus on dementia. We delineate the associations of subclinical dysfunction in specific organs with dementia risk and provide suggestions for public health promotion and clinical management.
Collapse
Affiliation(s)
- Eric E Smith
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, University of Calgary, Calgary, Canada.
| | - Geert Jan Biessels
- Department of Neurology, UMC Utrecht Brain Center, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Virginia Gao
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | | | - Arthur Liesz
- Institute for Stroke and Dementia Research, University Medical Center Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Neal S Parikh
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
2
|
Sümbül-Şekerci B, Pasin Ö, Egeli D, Gönenç S, Şekerci A. Characterizing cognitive phenotypes and clinical correlates in type 2 diabetes using fuzzy clustering and decision tree analysis. Sci Rep 2024; 14:23965. [PMID: 39397045 PMCID: PMC11471797 DOI: 10.1038/s41598-024-74741-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024] Open
Abstract
Cognitive impairment is frequently seen in patients with type 2 diabetes (T2DM), ranging from mild impairment to dementia. However, our knowledge of the specific profiles and risk factors for these different levels of impairment is limited. In this study involving 152 patients with T2DM, cognitive function was assessed using the Montreal Cognitive Assessment test. The Fuzzy C-means clustering algorithm was utilized to group individuals with similar cognitive characteristics. The study evaluated how well clinical parameters could classify characteristics of clusters using the Classification and Regression Trees algorithm. ROC analysis was then used to assess the classification success. Three distinct cognitive clusters were identified. Cluster 1 had the poorest cognitive performance and was characterized by more women, lower education levels, and lower levels of iron, hemoglobin, and creatine. Cluster 3, the amnestic cluster, was distinguished by low TSH levels. The decision tree model highlighted several parameters, including education level, hemoglobin, duration of diabetes mellitus (DM), iron, TSH, gender, family history of diabetes, and microalbumin/creatinine ratio, as significantly affecting the distinction of cognitive clusters. Diabetes-associated cognitive impairment stems from multifaceted pathophysiological mechanisms influenced by complex risk factors, resulting in diverse types of cognitive deficits.
Collapse
Affiliation(s)
- Betül Sümbül-Şekerci
- Faculty of Pharmacy, Clinical Pharmacy Department, Bezmialem Vakif University, Istanbul, Türkiye.
| | - Özge Pasin
- Faculty of Medicine, Biostatistics Department, Bezmialem Vakif University, Istanbul, Türkiye
| | - Derya Egeli
- Department of Pharmacognosy and Natural Product Chemistry Istanbul Institute of Health Sciences, Bezmialem Vakif University, Istanbul, Türkiye
| | - Senem Gönenç
- Faculty of Science, Statistics Department, Ataturk University, Erzurum, Türkiye
| | - Abdüsselam Şekerci
- Faculty of Medicine, Internal Medicine Department, Bezmialem Vakif University, Istanbul, Türkiye
| |
Collapse
|
3
|
Krizan I, Solingapuram Sai KK, Damuka N, Macauley SL, Maria Thurman B, Long M, Kavanagh K. Exploratory Dual PET imaging of [ 18F] fluorodeoxyglucose and [ 11C]acetoacetate in type 2 diabetic nonhuman primates. Bioorg Med Chem Lett 2024; 111:129906. [PMID: 39059565 PMCID: PMC11403582 DOI: 10.1016/j.bmcl.2024.129906] [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: 05/01/2024] [Revised: 07/20/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Despite recent advancements in imaging (amyloid-PET & tau-PET) and fluid (Aβ42/Aβ40 & Aβ42/ptau) biomarkers, the current standard for in vivo assessment of AD, diagnosis and prediction of Alzheimer's disease (AD) remains challenging. We demonstrated in nonhuman primates (NHP) that increased plasma and cerebrospinal fluid (CSF) glucose correlated with decreased CSF Aβ42 and CSF Aβ40, a hallmark of plaque promoting pathogenesis. Together, our findings demonstrate that altered glucose homeostasis and insulin resistance are associated with Aβ and amyloid in rodent and NHP models. This warranted further exploration into the dynamics of altered brain metabolism in the NHP model of T2D, cross referenced with CSF and blood-based AD markers. Preliminary dual PET ([11C]acetoacetate ([11C]AcAc) and [18F]fluorodeoxyglucose ([18F]FDG) imaging studies were conducted in an aged cohort of NHPs classified as T2D (n = 5) and pre-diabetic (n = 1) along with corresponding plasma and CSF samples for metabolite analysis. [11C]AcAc and [18F]FDG PET brain standard uptake values (SUV) were highly positively associated (r = 0.88, p = 0.02) in the T2D and pre-diabetic NHPs. Age was not significantly associated with brain SUV (age range 16.5-23.5 years old). Metabolic measures were positively correlated with brain [18F]FDG and CSF Aβ42:40 was positively correlated to fasting glucose values. Although our findings suggest moderate correlations, this study further elucidates that peripheral insulin resistance and poor glycemia control alter AD-related pathology, illustrating how T2D is a risk factor for AD.
Collapse
Affiliation(s)
- Ivan Krizan
- Department of Radiology, Wake Forest University School of Medicine, USA
| | | | - Naresh Damuka
- Department of Radiology, Wake Forest University School of Medicine, USA
| | - Shannon L Macauley
- Department of Physiology, University of Kentucky College of Medicine, USA
| | | | - Masha Long
- Department of Physiology, University of Kentucky College of Medicine, USA
| | - Kylie Kavanagh
- Department of Physiology, University of Kentucky College of Medicine, USA; College of Health and Medicine, University of Tasmania, Australia.
| |
Collapse
|
4
|
Sumbul‐Sekerci B, Velioglu HA, Sekerci A. Diabetes-related clinical and microstructural white matter changes in patients with Alzheimer's disease. Brain Behav 2024; 14:e3533. [PMID: 38715429 PMCID: PMC11077244 DOI: 10.1002/brb3.3533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 05/12/2024] Open
Abstract
AIM Although there exists substantial epidemiological evidence indicating an elevated risk of dementia in individuals with diabetes, our understanding of the neuropathological underpinnings of the association between Type-2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) remains unclear. This study aims to unveil the microstructural brain changes associated with T2DM in AD and identify the clinical variables contributing to these changes. METHODS In this retrospective study involving 64 patients with AD, 31 individuals had concurrent T2DM. The study involved a comparative analysis of diffusion tensor imaging (DTI) images and clinical features between patients with and without T2DM. The FSL FMRIB software library was used for comprehensive preprocessing and tractography analysis of DTI data. After eddy current correction, the "bedpost" model was utilized to model diffusion parameters. Linear regression analysis with a stepwise method was used to predict the clinical variables that could lead to microstructural white matter changes. RESULTS We observed a significant impairment in the left superior longitudinal fasciculus (SLF) among patients with AD who also had T2DM. This impairment in patients with AD and T2DM was associated with an elevation in creatine levels. CONCLUSION The white matter microstructure in the left SLF appears to be sensitive to the impairment of kidney function associated with T2DM in patients with AD. The emergence of AD in association with T2DM may be driven by mechanisms distinct from the typical AD pathology. Compromised renal function in AD could potentially contribute to impaired white matter integrity.
Collapse
Affiliation(s)
- Betul Sumbul‐Sekerci
- Department of Clinical Pharmacy, Faculty of PharmacyBezmialem Vakıf UniversityIstanbulTurkey
| | - Halil Aziz Velioglu
- Center for Psychiatric NeuroscienceFeinstein Institutes for Medical ResearchManhassetNew YorkUSA
- Department of Neuroscience, Faculty of MedicineIstanbul Medipol UniversityIstanbulTurkey
| | - Abdusselam Sekerci
- Department of Internal Medicine, Faculty of MedicineBezmialem Vakif UniversityIstanbulTurkey
| |
Collapse
|
5
|
Sood A, Capuano AW, Wilson RS, Barnes LL, Kapasi A, Bennett DA, Arvanitakis Z. Metformin, age-related cognitive decline, and brain pathology. Neurobiol Aging 2024; 133:99-106. [PMID: 37931533 PMCID: PMC10841359 DOI: 10.1016/j.neurobiolaging.2023.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 11/08/2023]
Abstract
The objective of this study was to evaluate the relation of metformin with change in cognition and brain pathology. During a mean of 8 years (SD = 5.5) of annual follow-up visits, 262/3029 participants were using metformin at any time during the study. Using a linear-mixed effect model adjusted for age, sex, and education, metformin users had slower decline on a score of global cognition compared to non-users (estimate = 0.017, SE = 0.007, p = 0.027). Analyses of cognitive domains showed a slower decline in episodic memory and semantic memory specifically. In sensitivity analysis, when examining any diabetes medication use vs none, no association was observed of any diabetes medication use with cognitive function. In the autopsy subset of 1584 participants, there was no difference in the level of Alzheimer's disease (AD) pathology or the presence of infarcts (of any size or location) between groups of metformin users vs non-users. However, in additional analyses, metformin users had higher odds of subcortical infarcts, and lower odds of atherosclerosis and arteriosclerosis.
Collapse
Affiliation(s)
- Ajay Sood
- Rush Alzheimer's Disease Center, Chicago, IL, USA.
| | | | | | | | | | | | | |
Collapse
|
6
|
Tahmi M, Benitez R, Luchsinger JA. Metformin as a Potential Prevention Strategy for Alzheimer's Disease and Alzheimer's Disease Related Dementias. J Alzheimers Dis 2024; 101:S345-S356. [PMID: 39422959 DOI: 10.3233/jad-240495] [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] [Indexed: 10/19/2024]
Abstract
Background Metformin is a safe and effective medication for type 2 diabetes (T2D) that has been proposed to decrease the risk of aging related disorders including Alzheimer's disease (AD) and Alzheimer's disease related disorders(ADRD). Objective This review seeks to summarize findings from studies examining the association of metformin with AD/ADRD related outcomes. Methods This is a narrative review of human studies, including observational studies and clinical trials, examining the association of metformin with cognitive and brain outcomes. We used PubMed as the main database for our literature search with a focus on English language human studies including observational studies and clinical trials. We prioritized studies published from 2013 until February 15, 2024. Results Observational human studies are conflicting, but those with better study designs suggest that metformin use in persons with T2D is associated with a lower risk of dementia. However, these observational studies are limited by the use of administrative data to ascertain metformin use and/or cognitive outcomes. There are few clinical trials in persons without T2D that have small sample sizes and short durations but suggest that metformin could prevent AD/ADRD. There are ongoing studies including large clinical trials with long duration that are testing the effect of metformin on AD/ADRD outcomes in persons without T2D at risk for dementia. Conclusions Clinical trial results are needed to establish the effect of metformin on the risk of AD and ADRD.
Collapse
Affiliation(s)
- Mouna Tahmi
- Department of Neurology, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Richard Benitez
- Departments of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - José A Luchsinger
- Departments of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
- Department of Epidemiology, Joseph P. Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
7
|
Santiago JA, Karthikeyan M, Lackey M, Villavicencio D, Potashkin JA. Diabetes: a tipping point in neurodegenerative diseases. Trends Mol Med 2023; 29:1029-1044. [PMID: 37827904 PMCID: PMC10844978 DOI: 10.1016/j.molmed.2023.09.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/11/2023] [Accepted: 09/21/2023] [Indexed: 10/14/2023]
Abstract
Diabetes is associated with an increased risk and progression of Alzheimer's (AD) and Parkinson's (PD) diseases. Conversely, diabetes may confer neuroprotection against amyotrophic lateral sclerosis (ALS). It has been posited that perturbations in glucose and insulin regulation, cholesterol metabolism, and mitochondrial bioenergetics defects may underlie the molecular underpinnings of diabetes effects on the brain. Nevertheless, the precise molecular mechanisms remain elusive. Here, we discuss the evidence from molecular, epidemiological, and clinical studies investigating the impact of diabetes on neurodegeneration and highlight shared dysregulated pathways between these complex comorbidities. We also discuss promising antidiabetic drugs, molecular diagnostics currently in clinical trials, and outstanding questions and challenges for future pursuit.
Collapse
Affiliation(s)
| | | | | | | | - Judith A Potashkin
- Center for Neurodegenerative Diseases and Therapeutics, Cellular and Molecular Pharmacology Department, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.
| |
Collapse
|
8
|
Kakraba S, Ayyadevara S, Mainali N, Balasubramaniam M, Bowroju S, Penthala NR, Atluri R, Barger SW, Griffin ST, Crooks PA, Shmookler Reis RJ. Thiadiazolidinone (TDZD) Analogs Inhibit Aggregation-Mediated Pathology in Diverse Neurodegeneration Models, and Extend C. elegans Life- and Healthspan. Pharmaceuticals (Basel) 2023; 16:1498. [PMID: 37895969 PMCID: PMC10610358 DOI: 10.3390/ph16101498] [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: 08/25/2023] [Revised: 09/29/2023] [Accepted: 10/02/2023] [Indexed: 10/29/2023] Open
Abstract
Chronic, low-grade inflammation has been implicated in aging and age-dependent conditions, including Alzheimer's disease, cardiomyopathy, and cancer. One of the age-associated processes underlying chronic inflammation is protein aggregation, which is implicated in neuroinflammation and a broad spectrum of neurodegenerative diseases such as Alzheimer's, Huntington's, and Parkinson's diseases. We screened a panel of bioactive thiadiazolidinones (TDZDs) from our in-house library for rescue of protein aggregation in human-cell and C. elegans models of neurodegeneration. Among the tested TDZD analogs, PNR886 and PNR962 were most effective, significantly reducing both the number and intensity of Alzheimer-like tau and amyloid aggregates in human cell-culture models of pathogenic aggregation. A C. elegans strain expressing human Aβ1-42 in muscle, leading to AD-like amyloidopathy, developed fewer and smaller aggregates after PNR886 or PNR962 treatment. Moreover, age-progressive paralysis was reduced 90% by PNR886 and 75% by PNR962, and "healthspan" (the median duration of spontaneous motility) was extended 29% and 62%, respectively. These TDZD analogs also extended wild-type C. elegans lifespan by 15-30% (p < 0.001), placing them among the most effective life-extension drugs. Because the lead drug in this family, TDZD-8, inhibits GSK3β, we used molecular-dynamic tools to assess whether these analogs may also target GSK3β. In silico modeling predicted that PNR886 or PNR962 would bind to the same allosteric pocket of inactive GSK3β as TDZD-8, employing the same pharmacophore but attaching with greater avidity. PNR886 and PNR962 are thus compelling candidate drugs for treatment of tau- and amyloid-associated neurodegenerative diseases such as AD, potentially also reducing all-cause mortality.
Collapse
Affiliation(s)
- Samuel Kakraba
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
| | - Srinivas Ayyadevara
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
- Central Arkansas Veterans Healthcare Service, Little Rock, AR 72205, USA
| | - Nirjal Mainali
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
| | - Meenakshisundaram Balasubramaniam
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
| | - Suresh Bowroju
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.B.); (N.R.P.); (P.A.C.)
| | - Narsimha Reddy Penthala
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.B.); (N.R.P.); (P.A.C.)
| | - Ramani Atluri
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
| | - Steven W. Barger
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
- Central Arkansas Veterans Healthcare Service, Little Rock, AR 72205, USA
| | - Sue T. Griffin
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
- Central Arkansas Veterans Healthcare Service, Little Rock, AR 72205, USA
| | - Peter A. Crooks
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (S.B.); (N.R.P.); (P.A.C.)
| | - Robert J. Shmookler Reis
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.M.); (M.B.); (R.A.); (S.W.B.); (S.T.G.)
- Central Arkansas Veterans Healthcare Service, Little Rock, AR 72205, USA
| |
Collapse
|
9
|
Husain KH, Sarhan SF, AlKhalifa HKAA, Buhasan A, Moin ASM, Butler AE. Dementia in Diabetes: The Role of Hypoglycemia. Int J Mol Sci 2023; 24:9846. [PMID: 37372995 DOI: 10.3390/ijms24129846] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Hypoglycemia, a common consequence of diabetes treatment, is associated with severe morbidity and mortality and has become a major barrier to intensifying antidiabetic therapy. Severe hypoglycemia, defined as abnormally low blood glucose requiring the assistance of another person, is associated with seizures and comas, but even mild hypoglycemia can cause troubling symptoms such as anxiety, palpitations, and confusion. Dementia generally refers to the loss of memory, language, problem-solving, and other cognitive functions, which can interfere with daily life, and there is growing evidence that diabetes is associated with an increased risk of both vascular and non-vascular dementia. Neuroglycopenia resulting from a hypoglycemic episode in diabetic patients can lead to the degeneration of brain cells, with a resultant cognitive decline, leading to dementia. In light of new evidence, a deeper understating of the relationship between hypoglycemia and dementia can help to inform and guide preventative strategies. In this review, we discuss the epidemiology of dementia among patients with diabetes, and the emerging mechanisms thought to underlie the association between hypoglycemia and dementia. Furthermore, we discuss the risks of various pharmacological therapies, emerging therapies to combat hypoglycemia-induced dementia, as well as risk minimization strategies.
Collapse
Affiliation(s)
- Khaled Hameed Husain
- School of Medicine, Royal College of Surgeons in Ireland, Busaiteen, Adliya 15503, Bahrain
| | - Saud Faisal Sarhan
- School of Medicine, Royal College of Surgeons in Ireland, Busaiteen, Adliya 15503, Bahrain
| | | | - Asal Buhasan
- School of Medicine, Royal College of Surgeons in Ireland, Busaiteen, Adliya 15503, Bahrain
| | - Abu Saleh Md Moin
- Research Department, Royal College of Surgeons in Ireland, Busaiteen, Adliya 15503, Bahrain
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Busaiteen, Adliya 15503, Bahrain
| |
Collapse
|
10
|
Li Z, Jiang Y, Long C, Peng Q, Yue R. The gut microbiota-astrocyte axis: Implications for type 2 diabetic cognitive dysfunction. CNS Neurosci Ther 2023; 29 Suppl 1:59-73. [PMID: 36601656 PMCID: PMC10314112 DOI: 10.1111/cns.14077] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/20/2022] [Accepted: 12/18/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Diabetic cognitive dysfunction (DCD) is one of the most insidious complications of type 2 diabetes mellitus, which can seriously affect the ability to self-monitoring of blood glucose and the quality of life in the elderly. Previous pathological studies of cognitive dysfunction have focused on neuronal dysfunction, characterized by extracellular beta-amyloid deposition and intracellular tau hyperphosphorylation. In recent years, astrocytes have been recognized as a potential therapeutic target for cognitive dysfunction and important participants in the central control of metabolism. The disorder of gut microbiota and their metabolites have been linked to a series of metabolic diseases such as diabetes mellitus. The imbalance of intestinal flora has the effect of promoting the occurrence and deterioration of several diabetes-related complications. Gut microbes and their metabolites can drive astrocyte activation. AIMS We reviewed the pathological progress of DCD related to the "gut microbiota-astrocyte" axis in terms of peripheral and central inflammation, intestinal and blood-brain barrier (BBB) dysfunction, systemic and brain energy metabolism disorders to deepen the pathological research progress of DCD and explore the potential therapeutic targets. CONCLUSION "Gut microbiota-astrocyte" axis, unique bidirectional crosstalk in the brain-gut axis, mediates the intermediate pathological process of neurocognitive dysfunction secondary to metabolic disorders in diabetes mellitus.
Collapse
Affiliation(s)
- Zi‐Han Li
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Ya‐Yi Jiang
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Cai‐Yi Long
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Qian Peng
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Ren‐Song Yue
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| |
Collapse
|
11
|
The common genes involved in the pathogenesis of Alzheimer's disease and type 2 diabetes and their implication for drug repositioning. Neuropharmacology 2023; 223:109327. [PMID: 36368623 DOI: 10.1016/j.neuropharm.2022.109327] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/28/2022] [Accepted: 10/31/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND The prevalences of Alzheimer's disease (AD) and type 2 diabetes (T2D) continuously increase with the aging of world population. Clinical and epidemiological studies indicate that T2D is an important risk factor for AD. However, the mechanisms underlying the linkage of the two disorders are still not fully elucidated. The aim of this study is to explore the molecular mechanisms of their comorbidity and potential drug targets for AD treatment. METHODS We first compiled comprehensive lists of genes associated with AD and T2D, respectively. Then, we investigated the signatures of the shared genes and screened for interactions between the hub genes. Subsequently, we used Autodock Vina to perform molecular docking to predict new drug candidates. Lastly, structure and dynamics of docking results were examined by molecular dynamics simulation to verify drug reliability. RESULTS We obtained 917 AD-associated genes, 631 T2D-associated genes and 175 shared genes between the two disorders for subsequent analyses. Functional analysis revealed that metabolic process, lipid and atherosclerosis, AMPK signaling pathway, insulin resistance, chemokines and cytokines were enriched in the shared genes. In addition, 50 central hub genes were identified, including IL6, TNF, INS, IL1B, AKT1, VEGFA, IL10, TP53, PTGS2, TLR4, and others. Finally, we predicted new drug candidates (verdoheme and stannsoporfin) that could be potentially used for AD treatment. CONCLUSIONS Our study confirmed that there are important shared genes and pathways between AD and T2D, which may provide clues to reveal the molecular mechanism underlying the pathophysiology of the two diseases and help us to discover novel drug candidates for the treatment of AD. The results may also provide clues into identification of new targets and strategies for prevention and therapy of T2D that predisposes to AD.
Collapse
|
12
|
Sumbul-Sekerci B, Sekerci A, Pasin O, Durmus E, Yuksel-Salduz ZI. Cognition and BDNF levels in prediabetes and diabetes: A mediation analysis of a cross-sectional study. Front Endocrinol (Lausanne) 2023; 14:1120127. [PMID: 36936159 PMCID: PMC10019820 DOI: 10.3389/fendo.2023.1120127] [Citation(s) in RCA: 3] [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: 12/09/2022] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
AIM Clinical and epidemiological studies suggest links between dementias and Type 2 diabetes (T2DM). The underlying mechanisms of diabetes-related cognitive impairment are largely unknown. This study aims to investigate the role of BDNF in cognitive impairment in prediabetes and T2DM. METHODS The study included 68 patients with prediabetes (preDM), 96 patients with T2DM, and 65 healthy controls. The cognitive function of the patients was evaluated with the Montreal Cognitive Assessment (MoCA) test and serum BDNF levels were measured by Elisa. The MoCA scores and BDNF levels were compared between diabetes groups after adjusting for age, gender, and education using ANCOVA. The role of BDNF in the diabetes-related cognitive impairment was investigated through mediation analysis. RESULTS Patients with T2DM had significantly lower cognitive performance, particularly in memory. Diabetes was found to be a predictor of both cognitive impairment and BDNF levels. A significant increase in serum BDNF levels was observed in patients with T2DM. However, the mediator role of BDNF in the pathology of cognitive impairment in diabetes was not determined. CONCLUSION Cognitive impairment is prevalent in patients with T2DM and should be included in routine screening for complications. The results of the mediation analysis suggest that although BDNF is a biomarker affected by T2DM and cognition, it does not play a mediator role between cognitive impairment and diabetes.
Collapse
Affiliation(s)
- Betul Sumbul-Sekerci
- Department of Clinical Pharmacy, Faculty of Pharmacy, Bezmialem Vakif University, Istanbul, Türkiye
- *Correspondence: Betul Sumbul-Sekerci, ;
| | - Abdusselam Sekerci
- Department of Internal Medicine, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Türkiye
| | - Ozge Pasin
- Department of Biostatistics, Faculty of Medicine, Bezmialem Vakif University, Istanbul, Türkiye
| | - Ezgi Durmus
- Department of Medical Biochemistry, Bezmialem Vakif University, Istanbul, Türkiye
- Health Sciences Institute, Bezmialem Vakif University, Istanbul, Türkiye
| | | |
Collapse
|
13
|
Toppala S, Ekblad LL, Viitanen M, Rinne JO, Jula A. Impaired Early Insulin Response to Glucose Load Predicts Episodic Memory Decline: A 10-Year Population-Based Cohort Follow-Up of 45-74-Year-Old Men and Women. J Alzheimers Dis 2023; 92:349-359. [PMID: 36744339 PMCID: PMC10041429 DOI: 10.3233/jad-220894] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Diabetes increases the risk for cognitive decline, but the mechanisms behind this association remain unknown. Impaired early insulin secretion in elderly men and insulin resistance, both of which are pathophysiological features of type 2 diabetes, have previously been linked to Alzheimer's disease. OBJECTIVE To examine if the early insulin response to oral glucose load predicts cognitive performance after 10 years in men and women aged 45-74 years. METHODS This study was based on a subpopulation of the Health 2000 Survey, a Finnish nationwide, population-based health examination study, and its follow-up, the Health 2011 Study. In total, 961 45-74-year-old individuals (mean age at baseline 55.6 years, 55.8% women) were examined. An oral glucose tolerance test was performed in 2001-2002, and early insulin response was defined as the ratio of the 30-min increment in insulin concentration to that of glucose concentration. Cognitive function was evaluated at baseline and follow-up with categorical verbal fluency, word-list learning, and word-list delayed recall. Statistical analyses were performed using multivariable linear models adjusted for age, sex, education, APOE&z.epsi;4 genotype, vascular risk factors including diabetes, and depressive symptoms. RESULTS A lower early insulin response to glucose load predicted lower performance (β: 0.21, p = 0.03) and greater decline (β: 0.19, p = 0.03) in the word-list delayed recall test. Baseline early insulin response did not predict verbal fluency or word-list learning (all p-values≥0.13). CONCLUSION Our results suggest that decreased early insulin secretion predicts episodic memory decline in middle-aged to elderly men and women.
Collapse
Affiliation(s)
- Sini Toppala
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland.,Kuopio City Home Care, Rehabilitation and Medical Services for Elderly, Kuopio, Finland
| | - Laura L Ekblad
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Matti Viitanen
- City of Turku, Welfare Division, Department of Geriatrics, Turku City Hospital and University of Turku, Turku, Finland.,Division of Clinical Geriatrics, NVS, Karolinska Institutet, Stockholm, Sweden
| | - Juha O Rinne
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland.,InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Antti Jula
- National Institute for Health and Welfare, Turku, Finland
| |
Collapse
|
14
|
McDonald TS, Lerskiatiphanich T, Woodruff TM, McCombe PA, Lee JD. Potential mechanisms to modify impaired glucose metabolism in neurodegenerative disorders. J Cereb Blood Flow Metab 2023; 43:26-43. [PMID: 36281012 PMCID: PMC9875350 DOI: 10.1177/0271678x221135061] [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: 12/16/2021] [Revised: 09/01/2022] [Accepted: 09/21/2022] [Indexed: 01/28/2023]
Abstract
Neurodegeneration refers to the selective and progressive loss-of-function and atrophy of neurons, and is present in disorders such as Alzheimer's, Huntington's, and Parkinson's disease. Although each disease presents with a unique pattern of neurodegeneration, and subsequent disease phenotype, increasing evidence implicates alterations in energy usage as a shared and core feature in the onset and progression of these disorders. Indeed, disturbances in energy metabolism may contribute to the vulnerability of neurons to apoptosis. In this review we will outline these disturbances in glucose metabolism, and how fatty acids are able to compensate for this impairment in energy production in neurodegenerative disorders. We will also highlight underlying mechanisms that could contribute to these alterations in energy metabolism. A greater understanding of these metabolism-neurodegeneration processes could lead to improved treatment options for neurodegenerative disease patients.
Collapse
Affiliation(s)
- Tanya S McDonald
- School of Biomedical Sciences, Faculty of Medicine, The
University of Queensland, St. Lucia, Australia
| | - Titaya Lerskiatiphanich
- School of Biomedical Sciences, Faculty of Medicine, The
University of Queensland, St. Lucia, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, The
University of Queensland, St. Lucia, Australia
- Queensland Brain Institute, The University of Queensland, St.
Lucia, Australia
| | - Pamela A McCombe
- Centre for Clinical Research, Faculty of Medicine, The
University of Queensland, St. Lucia, Australia
- Department of Neurology, Royal Brisbane & Women’s Hospital,
Herston, Australia
| | - John D Lee
- School of Biomedical Sciences, Faculty of Medicine, The
University of Queensland, St. Lucia, Australia
| |
Collapse
|
15
|
Kulminski AM, Feng F, Loiko E, Nazarian A, Loika Y, Culminskaya I. Prevailing Antagonistic Risks in Pleiotropic Associations with Alzheimer's Disease and Diabetes. J Alzheimers Dis 2023; 94:1121-1132. [PMID: 37355909 PMCID: PMC10666173 DOI: 10.3233/jad-230397] [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] [Indexed: 06/26/2023]
Abstract
BACKGROUND The lack of efficient preventive interventions against Alzheimer's disease (AD) calls for identifying efficient modifiable risk factors for AD. As diabetes shares many pathological processes with AD, including accumulation of amyloid plaques and neurofibrillary tangles, insulin resistance, and impaired glucose metabolism, diabetes is thought to be a potentially modifiable risk factor for AD. Mounting evidence suggests that links between AD and diabetes may be more complex than previously believed. OBJECTIVE To examine the pleiotropic architecture of AD and diabetes mellitus (DM). METHODS Univariate and pleiotropic analyses were performed following the discovery-replication strategy using individual-level data from 10 large-scale studies. RESULTS We report a potentially novel pleiotropic NOTCH2 gene, with a minor allele of rs5025718 associated with increased risks of both AD and DM. We confirm previously identified antagonistic associations of the same variants with the risks of AD and DM in the HLA and APOE gene clusters. We show multiple antagonistic associations of the same variants with AD and DM in the HLA cluster, which were not explained by the lead SNP in this cluster. Although the ɛ2 and ɛ4 alleles played a major role in the antagonistic associations with AD and DM in the APOE cluster, we identified non-overlapping SNPs in this cluster, which were adversely and beneficially associated with AD and DM independently of the ɛ2 and ɛ4 alleles. CONCLUSION This study emphasizes differences and similarities in the heterogeneous genetic architectures of AD and DM, which may differentiate the pathogenic mechanisms of these diseases.
Collapse
Affiliation(s)
- Alexander M Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, 27705, USA
| | - Fan Feng
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, 27705, USA
| | - Elena Loiko
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, 27705, USA
| | - Alireza Nazarian
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, 27705, USA
| | - Yury Loika
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, 27705, USA
| | - Irina Culminskaya
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, 27705, USA
| |
Collapse
|
16
|
Ruthirakuhan M, Cogo‐Moreira H, Swardfager W, Herrmann N, Lanctot KL, Black SE. Cardiovascular Risk Factors and Risk of Alzheimer Disease and Mortality: A Latent Class Approach. J Am Heart Assoc 2022; 12:e025724. [PMID: 36565204 PMCID: PMC9973600 DOI: 10.1161/jaha.122.025724] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background Cardiovascular risk factors co-occur with one another, and little is known about the extent of their clustering and risk of Alzheimer disease (AD). We identify groups of cardiovascular risk factors in cognitively normal individuals and investigate between-group differences in incident AD and death. Methods and Results Cognitively normal individuals were recruited from the National Alzheimer's Coordinator Center. A latent class analysis was conducted with hypertension, hypercholesterolemia, heart condition, stroke, smoking history, diabetes, and high body mass index. Between-group differences in the incidence of AD, mortality, and mortality-adjusted AD were investigated. This study included 12 412 cognitively normal individuals (average follow-up, 65 months). Three groups were identified: (1) low probabilities of cardiovascular risk factors (reference; N=5398 [43%]), (2) hypertension and hypercholesterolemia (vascular-dominant; N=5721 [46%]), and (3) hypertension, hypercholesterolemia, diabetes, and high body mass index (vascular-metabolic; N=1293 [10%]). Both vascular groups were significantly older, had more men, were slightly less educated, and were slightly more cognitively impaired than the reference group (all P<0.05). However, only the vascular-metabolic group had a significantly younger age of death compared with the reference group (84.3 versus 88.7 years, P<0.001). Only the vascular-dominant group had a greater incidence of AD (odds ratio [OR], 1.30; P<0.001) compared with the reference group. Mortality was greater in the vascular-dominant (OR, 3.26; P<0.001) and vascular-metabolic groups (OR, 1.84; P=0.02). Mortality-adjusted AD was greater in the vascular-dominant (OR, 1.54; P=0.02) and vascular-metabolic groups (OR, 1.46; P=0.04). Conclusions Three distinct cardiovascular risk factor groups were identified in cognitively normal elderly individuals. Only the vascular-dominant group was associated with a greater incidence of AD. Selective mortality may contribute to the attenuated association between the vascular-metabolic group and incident AD.
Collapse
Affiliation(s)
- Myuri Ruthirakuhan
- Hurvitz Brain Sciences Research ProgramSunnybrook Research InstituteTorontoOntarioCanada,University of TorontoTorontoOntarioCanada
| | - Hugo Cogo‐Moreira
- Department of Education, ICT and LearningOstfold University CollegeHaldenNorway,Universidade Federal de Sao PauloSao PauloBrazil
| | - Walter Swardfager
- Hurvitz Brain Sciences Research ProgramSunnybrook Research InstituteTorontoOntarioCanada,University of TorontoTorontoOntarioCanada
| | - Nathan Herrmann
- Hurvitz Brain Sciences Research ProgramSunnybrook Research InstituteTorontoOntarioCanada,University of TorontoTorontoOntarioCanada
| | - Krista L. Lanctot
- Hurvitz Brain Sciences Research ProgramSunnybrook Research InstituteTorontoOntarioCanada,University of TorontoTorontoOntarioCanada
| | - Sandra E. Black
- Hurvitz Brain Sciences Research ProgramSunnybrook Research InstituteTorontoOntarioCanada,University of TorontoTorontoOntarioCanada
| |
Collapse
|
17
|
Sümbül Şekerci B, Şekerci A, Gelişin Ö. Association between the peripheral blood parameters and Alzheimer’s disease in patients with type-2 diabetes mellitus: a case–control study. Int J Diabetes Dev Ctries 2022. [DOI: 10.1007/s13410-022-01135-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
18
|
Manzali SB, Yu E, Ravona-Springer R, Livny A, Golan S, Ouyang Y, Lesman-Segev O, Liu L, Ganmore I, Alkelai A, Gan-Or Z, Lin HM, Heymann A, Schnaider Beeri M, Greenbaum L. Alzheimer’s Disease Polygenic Risk Score Is Not Associated With Cognitive Decline Among Older Adults With Type 2 Diabetes. Front Aging Neurosci 2022; 14:853695. [PMID: 36110429 PMCID: PMC9468264 DOI: 10.3389/fnagi.2022.853695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectivesMultiple risk loci for late-onset Alzheimer’s disease (LOAD) have been identified. Type 2 diabetes (T2D) is a risk factor for cognitive decline, dementia and Alzheimer’s disease (AD). We investigated the association of polygenic risk score (PRS) for LOAD with overall cognitive functioning and longitudinal decline, among older adults with T2D.MethodsThe study included 1046 Jewish participants from the Israel Diabetes and Cognitive Decline (IDCD) study, aged ≥ 65 years, diagnosed with T2D, and cognitively normal at baseline. The PRS included variants from 26 LOAD associated loci (at genome-wide significance level), and was calculated with and without APOE. Outcome measures, assessed in 18 months intervals, were global cognition and the specific domains of episodic memory, attention/working memory, executive functions, and language/semantic categorization. Random coefficient models were used for analysis, adjusting for demographic variables, T2D-related characteristics, and cardiovascular factors. Additionally, in a subsample of 202 individuals, we analyzed the association of PRS with the volumes of total gray matter, frontal lobe, hippocampus, amygdala, and white matter hyperintensities. Last, the association of PRS with amyloid beta (Aβ) burden was examined in 44 participants who underwent an 18F-flutemetamol PET scan.ResultsThe PRS was not significantly associated with overall functioning or decline in global cognition or any of the specific cognitive domains. Similarly, following correction for multiple testing, there was no association with Aβ burden and other brain imaging phenotypes.ConclusionOur results suggest that the cumulative effect of LOAD susceptibility loci is not associated with a greater rate of cognitive decline in older adults with T2D, and other pathways may underlie this link.
Collapse
Affiliation(s)
- Sigalit B. Manzali
- Department of Pathology, Sheba Medical Center, Tel Hashomer, Israel
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Israel
| | - Eric Yu
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Ramit Ravona-Springer
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Israel
- Memory Clinic, Sheba Medical Center, Tel Hashomer, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Abigail Livny
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Diagnostic Imaging, Sheba Medical Center, Tel Hashomer, Israel
| | - Sapir Golan
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yuxia Ouyang
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Orit Lesman-Segev
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Israel
- Department of Diagnostic Imaging, Sheba Medical Center, Tel Hashomer, Israel
| | - Lang Liu
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Ithamar Ganmore
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Israel
- Memory Clinic, Sheba Medical Center, Tel Hashomer, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Anna Alkelai
- Institute for Genomic Medicine, Columbia University Medical Center, New York, NY, United States
| | - Ziv Gan-Or
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- Montreal Neurological Institute, McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Hung-Mo Lin
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Anthony Heymann
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Maccabi Healthcare Services, Tel Aviv, Israel
| | - Michal Schnaider Beeri
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Israel
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Lior Greenbaum
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Danek Gertner Institute of Human Genetics, Sheba Medical Center, Tel Hashomer, Israel
- *Correspondence: Lior Greenbaum,
| |
Collapse
|
19
|
Tsubaki H, Mendsaikhan A, Buyandelger U, Tooyama I, Walker DG. Localization of Thioredoxin-Interacting Protein in Aging and Alzheimer's Disease Brains. NEUROSCI 2022; 3:166-185. [PMID: 39483368 PMCID: PMC11523753 DOI: 10.3390/neurosci3020013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 03/28/2022] [Indexed: 11/03/2024] Open
Abstract
Thioredoxin-Interacting Protein (TXNIP) has been shown to have significant pathogenic roles in many human diseases, particularly those associated with diabetes and hyperglycemia. Its main mode of action is to sequester thioredoxins, resulting in enhanced oxidative stress. The aim of this study was to identify if cellular expression of TXNIP in human aged and Alzheimer's disease (AD) brains correlated with pathological structures. This study employed fixed tissue sections and protein extracts of temporal cortex from AD and aged control brains. Studies employed light and fluorescent immunohistochemical techniques using the monoclonal antibody JY2 to TXNIP to identify cellular structures. Immunoblots were used to quantify relative amounts of TXNIP in brain protein extracts. The major finding was the identification of TXNIP immunoreactivity in selective neuronal populations and structures, particularly in non-AD brains. In AD brains, less neuronal TXNIP but increased numbers of TXNIP-positive plaque-associated microglia were observed. Immunoblot analyses showed no significant increase in levels of TXNIP protein in the AD samples tested. In conclusion, this study identified altered patterns of expression of TXNIP in human brains with progression of AD pathology.
Collapse
Affiliation(s)
- Haruka Tsubaki
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu 520-2192, Japan; (H.T.); (A.M.); (U.B.); (I.T.)
| | - Anarmaa Mendsaikhan
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu 520-2192, Japan; (H.T.); (A.M.); (U.B.); (I.T.)
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Undral Buyandelger
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu 520-2192, Japan; (H.T.); (A.M.); (U.B.); (I.T.)
| | - Ikuo Tooyama
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu 520-2192, Japan; (H.T.); (A.M.); (U.B.); (I.T.)
| | - Douglas G Walker
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu 520-2192, Japan; (H.T.); (A.M.); (U.B.); (I.T.)
- Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ 85281, USA
| |
Collapse
|
20
|
Honea RA, John CS, Green ZD, Kueck PJ, Taylor MK, Lepping RJ, Townley R, Vidoni ED, Burns JM, Morris JK. Relationship of fasting glucose and longitudinal Alzheimer's disease imaging markers. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12239. [PMID: 35128029 PMCID: PMC8804928 DOI: 10.1002/trc2.12239] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 11/30/2021] [Accepted: 12/15/2021] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Fasting glucose increases with age and is linked to modifiable Alzheimer's disease risk factors such as cardiovascular disease and Type 2 diabetes (T2D). METHODS We leveraged available biospecimens and neuroimaging measures collected during the Alzheimer's Prevention Through Exercise (APEx) trial (n = 105) to examine the longitudinal relationship between change in blood glucose metabolism and change in regional cerebral amyloid deposition and gray and white matter (WM) neurodegeneration in older adults over 1 year of follow-up. RESULTS Individuals with improving fasting glucose (n = 61) exhibited less atrophy and regional amyloid accumulation compared to those whose fasting glucose worsened over 1 year (n = 44). Specifically, while individuals with increasing fasting glucose did not yet show cognitive decline, they did have regional atrophy in the hippocampus and inferior parietal cortex, and increased amyloid accumulation in the precuneus cortex. Signs of early dementia pathology occurred in the absence of significant group differences in insulin or body composition, and was not modified by apolipoprotein E ε4 carrier status. DISCUSSION Dysregulation of glucose in late life may signal preclinical brain change prior to clinically relevant cognitive decline. Additional work is needed to determine whether treatments specifically targeting fasting glucose levels may impact change in brain structure or cerebral amyloid in older adults.
Collapse
Affiliation(s)
- Robyn A. Honea
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
- Department of NeurologyUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Casey S. John
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
- Department of NeurologyUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Zachary D. Green
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
- Department of NeurologyUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Paul J. Kueck
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
- Department of NeurologyUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Matthew K. Taylor
- Department of Dietetics and NutritionUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Rebecca J. Lepping
- Hoglund Biomedical Imaging CenterUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Ryan Townley
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
- Department of NeurologyUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Eric D. Vidoni
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
- Department of NeurologyUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Jeffery M. Burns
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
- Department of NeurologyUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Jill K. Morris
- University of Kansas Alzheimer's Disease Research CenterKansas CityKansasUSA
- Department of NeurologyUniversity of Kansas Medical CenterKansas CityKansasUSA
| |
Collapse
|
21
|
Zhang W, Lu J, Qing Z, Zhang X, Zhao H, Bi Y, Zhang B. Effects of Subcortical Atrophy and Alzheimer’s Pathology on Cognition in Elderly Type 2 Diabetes: The Alzheimer’s Disease Neuroimaging Initiative Study. Front Aging Neurosci 2022; 14:781938. [PMID: 35173604 PMCID: PMC8841716 DOI: 10.3389/fnagi.2022.781938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/05/2022] [Indexed: 12/03/2022] Open
Abstract
Background Subcortical atrophy and increased cerebral β-amyloid and tau deposition are linked to cognitive decline in type 2 diabetes. However, whether and how subcortical atrophy is related to Alzheimer’s pathology in diabetes remains unclear. This study therefore aimed to investigate subcortical structural alterations induced by diabetes and the relationship between subcortical alteration, Alzheimer’s pathology and cognition. Methods Participants were 150 patients with type 2 diabetes and 598 propensity score-matched controls without diabetes from the Alzheimer’s Disease Neuroimaging Initiative. All subjects underwent cognitive assessments, magnetic resonance imaging (MRI), and apolipoprotein E (ApoE) genotyping, with a subset that underwent amyloid positron emission tomography (PET) and cerebrospinal fluid (CSF) assays to determine cerebral β-amyloid deposition (n = 337) and CSF p-tau (n = 433). Subcortical structures were clustered into five modules based on Pearson’s correlation coefficients of volumes across all subjects: the ventricular system, the corpus callosum, the limbic system, the diencephalon, and the striatum. Using structural equation modeling (SEM), we investigated the relationships among type 2 diabetes, subcortical structural alterations, and AD pathology. Results Compared with the controls, the diabetic patients had significant reductions in the diencephalon and limbic system volumes; moreover, patients with longer disease duration (>6 years) had more severe volume deficit in the diencephalon. SEM suggested that type 2 diabetes, age, and the ApoE ε4 allele (ApoE-ε4) can affect cognition via reduced subcortical structure volumes (total effect: age > ApoE-ε4 > type 2 diabetes). Among them, age and ApoE-ε4 strongly contributed to AD pathology, while type 2 diabetes neither directly nor indirectly affected AD biomarkers. Conclusion Our study suggested the subcortical atrophy mediated the association of type 2 diabetes and cognitive decline. Although both type 2 diabetes and AD are correlated with subcortical neurodegeneration, type 2 diabetes have no direct or indirect effect on the cerebral amyloid deposition and CSF p-tau.
Collapse
Affiliation(s)
- Wen Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jiaming Lu
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhao Qing
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xin Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Hui Zhao
- Department of Neurology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yan Bi
- Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Bing Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- *Correspondence: Bing Zhang,
| | | |
Collapse
|
22
|
Sadrolashrafi K, Craft S, Decourt B, Adem A, Wilson JR, Miller J, Sabbagh MN. Is diabetes associated with increased pathological burden in Alzheimer's disease? ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12248. [PMID: 34796262 PMCID: PMC8579894 DOI: 10.1002/dad2.12248] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/04/2021] [Accepted: 08/31/2021] [Indexed: 12/17/2022]
Abstract
INTRODUCTION We examined the association between Alzheimer's disease (AD) and type 2 diabetes mellitus (DM) and hypothesized that diabetes is associated with an increased pathological burden in clinically and pathologically diagnosed AD. METHODS All data were obtained from the Uniform Data Set (UDS) v3, the Neuropathology Data Set, and the Researcher's Data Dictionary-Genetic Data from the National Alzheimer's Coordinating Center. The dataset (37 cases with diabetes and 1158 cases without) relies on autopsy-confirmed data in clinically diagnosed AD patients who were assessed for diabetes type in form A5 or D2 during at least one visit. Differences in scores were explored using a general linear model. Effect sizes were calculated using sample means and standard deviations (Cohen's d). RESULTS The presence of diabetes was associated with a lower Thal phase of amyloid plaques (A score; 4.6 ± 0.79 vs. 4.3 ± 0.85, P < .05) and lower Braak stage for neurofibrillary degeneration (B score; 5.58 ± 0.72 vs. 5.16 ± 0.96, P < 0.05) but not for density of neocortical neuritic plaques (CERAD score-C score). The National Institute on Aging-Alzheimer's Association Alzheimer's disease neuropathologic change (ABC score) was not different between AD+DM and AD-DM. DISCUSSION This pilot study found a significantly lower Thal phase of amyloid plaques and Braak stage for neurofibrillary degeneration in AD-confirmed individuals with diabetes compared to those without. Thus type 2 DM is not associated with increased AD pathology in clinically and pathologically confirmed cases of AD.
Collapse
Affiliation(s)
- Kaviyon Sadrolashrafi
- Cleveland Clinic Lou Ruvo Center for Brain HealthLas VegasNevadaUSA
- Kirk Kerkorian School of Medicineat University of Nevada, Las VegasLas VegasNevadaUSA
| | - Suzanne Craft
- Wake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Boris Decourt
- Cleveland Clinic Lou Ruvo Center for Brain HealthLas VegasNevadaUSA
| | - Abdu Adem
- United Arab Emirates UniversityAl‐AinUnited Arab Emirates
| | - Jeffrey R. Wilson
- Arizona State University W. P. Carey School of BusinessTempeArizonaUSA
| | - Justin Miller
- Cleveland Clinic Lou Ruvo Center for Brain HealthLas VegasNevadaUSA
| | - Marwan N. Sabbagh
- Cleveland Clinic Lou Ruvo Center for Brain HealthLas VegasNevadaUSA
- Barrow Neurological InstitutePhoenixAZ
| |
Collapse
|
23
|
Al-Janahi E, Ponirakis G, Al Hamad H, Vattoth S, Elsotouhy A, Petropoulos IN, Khan A, Gad H, Chandran M, Sankaranarayanan A, Ramadan M, Elorrabi M, Gadelseed M, Tosino R, Gawhale PV, Arasn A, Alobaidi M, Khan S, Manikoth P, Hamdi Y, Osman S, Nadukkandiyil N, AlSulaiti E, Thodi N, Almuhannadi H, Mahfoud ZR, Own A, Shuaib A, Malik RA. Corneal Nerve and Brain Imaging in Mild Cognitive Impairment and Dementia. J Alzheimers Dis 2021; 77:1533-1543. [PMID: 32925064 PMCID: PMC7683060 DOI: 10.3233/jad-200678] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background: Visual rating of medial temporal lobe atrophy (MTA) is an accepted structural neuroimaging marker of Alzheimer’s disease. Corneal confocal microscopy (CCM) is a non-invasive ophthalmic technique that detects neuronal loss in peripheral and central neurodegenerative disorders. Objective: To determine the diagnostic accuracy of CCM for mild cognitive impairment (MCI) and dementia compared to medial temporal lobe atrophy (MTA) rating on MRI. Methods: Subjects aged 60–85 with no cognitive impairment (NCI), MCI, and dementia based on the ICD-10 criteria were recruited. Subjects underwent cognitive screening, CCM, and MTA rating on MRI. Results: 182 subjects with NCI (n = 36), MCI (n = 80), and dementia (n = 66), including AD (n = 19, 28.8%), VaD (n = 13, 19.7%), and mixed AD (n = 34, 51.5%) were studied. CCM showed a progressive reduction in corneal nerve fiber density (CNFD, fibers/mm2) (32.0±7.5 versus 24.5±9.6 and 20.8±9.3, p < 0.0001), branch density (CNBD, branches/mm2) (90.9±46.5 versus 59.3±35.7 and 53.9±38.7, p < 0.0001), and fiber length (CNFL, mm/mm2) (22.9±6.1 versus 17.2±6.5 and 15.8±7.4, p < 0.0001) in subjects with MCI and dementia compared to NCI. The area under the ROC curve (95% CI) for the diagnostic accuracy of CNFD, CNBD, CNFL compared to MTA-right and MTA-left for MCI was 78% (67–90%), 82% (72–92%), 86% (77–95%) versus 53% (36–69%) and 40% (25–55%), respectively, and for dementia it was 85% (76–94%), 84% (75–93%), 85% (76–94%) versus 86% (76–96%) and 82% (72–92%), respectively. Conclusion: The diagnostic accuracy of CCM, a non-invasive ophthalmic biomarker of neurodegeneration, was high and comparable with MTA rating for dementia but was superior to MTA rating for MCI.
Collapse
Affiliation(s)
- Eiman Al-Janahi
- Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - Georgios Ponirakis
- Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, Doha, Qatar.,Manchester Metropolitan University, Faculty of Science and Engineering, Manchester, UK
| | - Hanadi Al Hamad
- Geriatric & Memory Clinic, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Surjith Vattoth
- Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, Doha, Qatar.,Neuroradiology, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Ahmed Elsotouhy
- Neuroradiology, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| | | | - Adnan Khan
- Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - Hoda Gad
- Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - Mani Chandran
- Geriatric & Memory Clinic, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
| | | | - Marwan Ramadan
- Geriatric & Memory Clinic, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Marwa Elorrabi
- Geriatric & Memory Clinic, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Masharig Gadelseed
- Geriatric & Memory Clinic, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Rhia Tosino
- Geriatric & Memory Clinic, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Priya V Gawhale
- Geriatric & Memory Clinic, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Anjum Arasn
- Geriatric & Memory Clinic, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Maryam Alobaidi
- Geriatric & Memory Clinic, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Shafi Khan
- Geriatric & Memory Clinic, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Pravija Manikoth
- Geriatric & Memory Clinic, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Yasmin Hamdi
- Geriatric & Memory Clinic, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Susan Osman
- Geriatric & Memory Clinic, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Navas Nadukkandiyil
- Geriatric & Memory Clinic, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Essa AlSulaiti
- Geriatric & Memory Clinic, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Noushad Thodi
- MRI Unit, Rumailah Hospital, Hamad Medical Corporation, Doha, Qatar
| | - Hamad Almuhannadi
- Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - Ziyad R Mahfoud
- Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - Ahmed Own
- School of Medicine, Western Sydney University, New South Wales, Australia
| | - Ashfaq Shuaib
- Department of Medicine, University of Alberta, Alberta, Canada
| | - Rayaz A Malik
- Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, Doha, Qatar.,Manchester Metropolitan University, Faculty of Science and Engineering, Manchester, UK.,Institute of Cardiovascular Science, University of Manchester, Manchester, UK
| |
Collapse
|
24
|
Fajersztajn L, Di Rienzo V, Nakamura CA, Scazufca M. Watching TV and Cognition: The SPAH 2-Year Cohort Study of Older Adults Living in Low-Income Communities. Front Neurol 2021; 12:628489. [PMID: 34248811 PMCID: PMC8267001 DOI: 10.3389/fneur.2021.628489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 06/02/2021] [Indexed: 11/13/2022] Open
Abstract
Watching TV is a highly prevalent leisure activity among older adults and, in many cases, the only leisure option of those living in low-income communities. While engaging in leisure activities have proven to protect older adults from cognitive decline, the effects of watching TV on cognition of this population is controversial in the literature. This study investigated the impact of watching TV on global cognitive function, immediate memory, verbal fluency, risk of dementia of amnestic mild cognitive impairment (aMCI) in a cohort of older adults residents of socioeconomically deprived areas of São Paulo, Brazil. We used data from the São Paulo Aging & Health Study (SPAH). Participants aged 65 years or over, with no dementia diagnosis at baseline and who completed the 2-year follow-up assessment were included in this study (n = 1,243). Multivariable linear regression models were performed to assess the effect of watching TV on global cognitive function, immediate memory and verbal fluency. Multivariable logistic regression models were used to evaluate the risk of developing dementia and aMCI. Models were controlled by cognitive performance at baseline, sociodemographic characteristics and functional status. Cognitive performance at baseline and follow-up were similar. Thirty-one participants were diagnosed with dementia, and 23 with aMCI 24 months after inclusion in the study. Watching TV did not show any positive or negative effect on global cognitive function, immediate memory, verbal fluency, risk of dementia and risk of aMCI. It is good news that watching TV did not predict the decline in cognition in elders. However, it is essential to increase opportunities for other leisure activities for low-income and low-educated older adults if we do consider that leisure activities protect cognition decline in older adults. In the coming decades, developing countries will experience the highest burden of dementia and more than fun, public policies to promote leisure activities might be a strategy to alleviate this burden shortly.
Collapse
Affiliation(s)
- Laís Fajersztajn
- Laboratório de Poluição Ambiental, Departamento de Patologia, Faculdade de Medicina (FMUSP), Universidade de São Paulo, São Paulo, Brazil
| | - Vanessa Di Rienzo
- Faculdade de Medicina (FMUSP), Universidade de São Paulo, São Paulo, Brazil.,Universidade São Judas Tadeu, São Paulo, Brazil
| | - Carina Akemi Nakamura
- Faculdade de Medicina (FMUSP), Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Investigação Medica (LIM) 23, Faculdade de Medicina, Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Universidade de São Paulo, São Paulo, Brazil
| | - Marcia Scazufca
- Faculdade de Medicina (FMUSP), Universidade de São Paulo, São Paulo, Brazil.,Laboratório de Investigação Medica (LIM) 23, Faculdade de Medicina, Instituto de Psiquiatria, Hospital das Clinicas HCFMUSP, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
25
|
Tanokashira D, Wang W, Maruyama M, Kuroiwa C, White MF, Taguchi A. Irs2 deficiency alters hippocampus-associated behaviors during young adulthood. Biochem Biophys Res Commun 2021; 559:148-154. [PMID: 33940386 DOI: 10.1016/j.bbrc.2021.04.101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 04/24/2021] [Indexed: 10/21/2022]
Abstract
Type 2 diabetes mellitus (T2DM), characterized by hyperglycemia and insulin resistance, has been recognized as a risk factor for cognitive impairment and dementia, including Alzheimer's disease (AD). Insulin receptor substrate2 (IRS2) is a major component of the insulin/insulin-like growth factor-1 signaling pathway. Irs2 deletion leads to life-threatening T2DM, promoting premature death in male mice regardless of their genetic background. Here, we showed for the first time that young adult male mice lacking Irs2 on a C57BL/6J genetic background (Irs2-/-/6J) survived in different experimental environments and displayed hippocampus-associated behavioral alterations. Young adult male Irs2-/-/6J mice also exhibit aberrant alterations in energy and nutrient sensors, such as AMP-activated protein kinase (AMPK) and glucose transporter3 (GLUT3), and reduced core body temperature accompanied by abnormal change in the temperature sensor in the brain. These results suggest that Irs2 deficiency-induced impairments of brain energy metabolism and thermoregulation contribute to hippocampus-associated behavioral changes in young adult male mice.
Collapse
Affiliation(s)
- Daisuke Tanokashira
- Department of Integrative Neuroscience, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
| | - Wei Wang
- Department of Integrative Neuroscience, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
| | - Megumi Maruyama
- Department of Integrative Neuroscience, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
| | - Chiemi Kuroiwa
- Department of Integrative Neuroscience, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan
| | - Morris F White
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Akiko Taguchi
- Department of Integrative Neuroscience, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan.
| |
Collapse
|
26
|
Shinohara M, Kikuchi M, Onishi-Takeya M, Tashiro Y, Suzuki K, Noda Y, Takeda S, Mukouzono M, Nagano S, Fukumori A, Morishita R, Nakaya A, Sato N. Upregulated expression of a subset of genes in APP; ob/ ob mice: Evidence of an interaction between diabetes-linked obesity and Alzheimer's disease. FASEB Bioadv 2021; 3:323-333. [PMID: 33977233 PMCID: PMC8103720 DOI: 10.1096/fba.2020-00151] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/17/2021] [Accepted: 01/29/2021] [Indexed: 12/26/2022] Open
Abstract
Clinical studies have indicated that obesity and diabetes are associated with Alzheimer's disease (AD) and neurodegeneration. However, the mechanism by which obesity/diabetes and AD interact with each other and contribute to dementia remains elusive. To obtain insights into their interaction at molecular levels, we performed gene expression analysis of APP;ob/ob mice, which were generated by crossing transgenic AD model mice (APP23 mice) with ob/ob mice, which are obese and mildly diabetic. The Aβ level in these mice was reduced compared with that in pure APP mice. However, we identified a cluster of genes (cluster 10) upregulated in APP;ob/ob mice but not in either APP or ob/ob mice. Interestingly, genes upregulated in the human AD brain were enriched in cluster 10. Moreover, genes in cluster 10 formed a network and shared upregulated genes with a cell model of neurodegeneration and other models of neurological disorders such as ischemia and epilepsy. In silico analyses showed that serum response factor (SRF), recently identified in a single-cell analysis of human brains as a transcription factor that can control the conversion from healthy cells to AD cells, might be a common transcriptional regulator for a subset of cluster 10 genes. These data suggest that upregulation of genes uniquely associated with APP;ob/ob mice is an evidence of the interaction between obesity/diabetes and AD.
Collapse
Affiliation(s)
- Mitsuru Shinohara
- Department of Aging Neurobiology Center for Development of Advanced Medicine for Dementia National Center for Geriatrics and Gerontology Obu Aichi Japan.,Department of Aging Neurobiology Graduate School of Medicine Osaka University Osaka Japan
| | - Masataka Kikuchi
- Department of Genome Informatics Graduate School of Medicine Osaka University Osaka Japan
| | - Miyuki Onishi-Takeya
- Department of Geriatric Medicine Graduate School of Medicine Osaka University Osaka Japan
| | - Yoshitaka Tashiro
- Department of Aging Neurobiology Center for Development of Advanced Medicine for Dementia National Center for Geriatrics and Gerontology Obu Aichi Japan
| | - Kaoru Suzuki
- Department of Aging Neurobiology Center for Development of Advanced Medicine for Dementia National Center for Geriatrics and Gerontology Obu Aichi Japan
| | - Yasuhiro Noda
- Department of Aging Neurobiology Center for Development of Advanced Medicine for Dementia National Center for Geriatrics and Gerontology Obu Aichi Japan
| | - Shuko Takeda
- Department of Clinical Gene Therapy Graduate School of Medicine Osaka University Osaka Japan
| | - Masahiro Mukouzono
- Department of Clinical Gene Therapy Graduate School of Medicine Osaka University Osaka Japan
| | - Seiichi Nagano
- Department of Neurology Graduate School of Medicine Osaka University Osaka Japan
| | - Akio Fukumori
- Department of Aging Neurobiology Center for Development of Advanced Medicine for Dementia National Center for Geriatrics and Gerontology Obu Aichi Japan.,Department of Aging Neurobiology Graduate School of Medicine Osaka University Osaka Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy Graduate School of Medicine Osaka University Osaka Japan
| | - Akihiro Nakaya
- Department of Genome Informatics Graduate School of Medicine Osaka University Osaka Japan
| | - Naoyuki Sato
- Department of Aging Neurobiology Center for Development of Advanced Medicine for Dementia National Center for Geriatrics and Gerontology Obu Aichi Japan.,Department of Aging Neurobiology Graduate School of Medicine Osaka University Osaka Japan
| |
Collapse
|
27
|
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
|
28
|
Increase in Number of Depression Symptoms Over Time is Related to Worse Cognitive Outcomes in Older Adults With Type 2 Diabetes. Am J Geriatr Psychiatry 2021; 29:1-11. [PMID: 33127316 PMCID: PMC7771631 DOI: 10.1016/j.jagp.2020.09.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 11/24/2022]
Abstract
OBJECTIVE Older adults with type 2 diabetes (T2D) are at increased risk for depression, cognitive decline, and dementia compared to those without T2D. Little is known about the association of simultaneous changes in depression symptoms and cognitive decline over time. METHODS Subjects (n=1021; mean age 71.6 [SD=4.6]; 41.2% female) were initially cognitively normal participants of the Israel Diabetes and Cognitive Decline study who underwent evaluations of depression and cognition approximately every 18 months. Cognitive tests were summarized into four cognitive domains: episodic memory, attention/working memory, executive functions, and semantic categorization. The average of the z-scores of the four domains defined global cognition. Depression symptoms were assessed using the Geriatric Depression Scale, 15-item version. We fit a random coefficients model of changes in depression and in cognitive functions, adjusting for baseline sociodemographic and cardiovascular variables. RESULTS Higher number of depression symptoms at baseline was significantly associated with lower baseline cognitive scores in global cognition (estimate = -0.1175, SE = 0.021, DF = 1,014, t = -5.59; p < 0.001), executive functions (estimate = -0.186, SE = 0.036, DF = 1,013, t = -5.15; p = <0.001), semantic categorization (estimate = -0.155, SE = 0.029, DF = 1,008, t = -5.3; p < 0.001), and episodic memory (estimate = -0.08165, SE = 0.027, DF = 1,035, t = -2.92; p = 0.0036), but not with rate of decline in any cognitive domain. During follow-up, a larger increase in number of depression symptoms, was associated with worse cognitive outcomes in global cognition (estimate = -0.1053, SE = 0.027, DF = 1,612, t = -3.77; p = 0.0002), semantic categorization (estimate = -0.123, SE = 0.036, DF = 1,583, t = -3.36; p = 0.0008), and in episodic memory (estimate = -0.165, SE = 0.055, DF = 1,622, t = -3.02; p = 0.003), but the size of this effect was constant over time. CONCLUSION In elderly with T2D, increase in depression symptoms over time is associated with parallel cognitive decline, indicating that the natural course of the two conditions progresses concurrently and suggesting common underlying mechanisms".
Collapse
|
29
|
Barthold D, Gibbons LE, Marcum ZA, Gray SL, Keene CD, Grabowski TJ, Postupna N, Larson EB, Crane PK. Alzheimer's Disease-Related Neuropathology Among Patients with Medication Treated Type 2 Diabetes in a Community-Based Autopsy Cohort. J Alzheimers Dis 2021; 83:1303-1312. [PMID: 34420950 PMCID: PMC8846930 DOI: 10.3233/jad-210059] [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] [Indexed: 01/18/2023]
Abstract
BACKGROUND Diabetes is a risk factor for Alzheimer's disease and related dementias (ADRD). Epidemiologic evidence shows an association between diabetes medications and ADRD risk; cell and mouse models show diabetes medication association with AD-related neuropathologic change (ADNC). OBJECTIVE This hypothesis-generating analysis aimed to describe autopsy-measured ADNC for individuals who used diabetes medications. METHODS Descriptive analysis of ADNC for Adult Changes in Thought (ACT) Study autopsy cohort who used diabetes medications, including sulfonylureas, insulin, and biguanides; total N = 118. ADNC included amyloid plaque distribution (Thal phasing), neurofibrillary tangle (NFT) distribution (Braak stage), and cortical neuritic plaque density (CERAD score). We also examined quantitative measures of ADNC using the means of standardized Histelide measures of cortical PHF-tau and Aβ1-42. Adjusted analyses control for age at death, sex, education, APOE genotype, and diabetes complication severity index. RESULTS Adjusted analyses showed no significant association between any drug class and traditional neuropathologic measures compared to nonusers of that class. In adjusted Histelide analyses, any insulin use was associated with lower mean levels of Aβ1-42 (-0.57 (CI: -1.12, -0.02)) compared to nonusers. Five years of sulfonylureas and of biguanides use was associated with lower levels of Aβ1-42 compared to nonusers (-0.15 (CI: -0.28, -0.02), -0.31 (CI: -0.54, -0.07), respectively). CONCLUSION Some evidence exists that diabetes medications are associated with lower levels of Aβ1-42, but not traditional measures of neuropathology. Future studies are needed in larger samples to build understanding of the mechanisms between diabetes, its medications, and ADRD, and to potentially repurpose existing medications for prevention or delay of ADRD.
Collapse
Affiliation(s)
- Douglas Barthold
- The Comparative Health Outcomes, Policy, and Economics (CHOICE) Institute, Department of Pharmacy, University of Washington, Seattle, WA, USA
- The Plein Center for Geriatric Pharmacy Research, Education, and Outreach, School of Pharmacy, University of Washington, Seattle, WA, USA
| | - Laura E. Gibbons
- General Internal Medicine, Data Management and Statistics Core, Alzheimer’s Disease Research Center, University of Washington, Seattle, WA, USA
- Department of Medicine, UW School of Medicine, University of Washington, Seattle, WA, USA
| | - Zachary A. Marcum
- The Comparative Health Outcomes, Policy, and Economics (CHOICE) Institute, Department of Pharmacy, University of Washington, Seattle, WA, USA
- The Plein Center for Geriatric Pharmacy Research, Education, and Outreach, School of Pharmacy, University of Washington, Seattle, WA, USA
| | - Shelly L. Gray
- The Comparative Health Outcomes, Policy, and Economics (CHOICE) Institute, Department of Pharmacy, University of Washington, Seattle, WA, USA
- The Plein Center for Geriatric Pharmacy Research, Education, and Outreach, School of Pharmacy, University of Washington, Seattle, WA, USA
| | - C. Dirk Keene
- Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, USA
| | - Thomas J. Grabowski
- Department of Radiology and Neurology, UW School of Medicine, Alzheimer’s Disease Research Center, University of Washington, Seattle, WA, USA
| | - Nadia Postupna
- Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, USA
| | - Eric B. Larson
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
- Department of Medicine, UW School of Medicine, University of Washington, Seattle, WA, USA
| | - Paul K. Crane
- Department of Medicine, UW School of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
30
|
Toppala S, Ekblad LL, Lötjönen J, Helin S, Hurme S, Johansson J, Jula A, Karrasch M, Koikkalainen J, Laine H, Parkkola R, Viitanen M, Rinne JO. Midlife Insulin Resistance as a Predictor for Late-Life Cognitive Function and Cerebrovascular Lesions. J Alzheimers Dis 2020; 72:215-228. [PMID: 31561373 PMCID: PMC6839606 DOI: 10.3233/jad-190691] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background: Type 2 diabetes (T2DM) increases the risk for Alzheimer’s disease (AD) but not for AD neuropathology. The association between T2DM and AD is assumed to be mediated through vascular mechanisms. However, insulin resistance (IR), the hallmark of T2DM, has been shown to associate with AD neuropathology and cognitive decline. Objective: To evaluate if midlife IR predicts late-life cognitive performance and cerebrovascular lesions (white matter hyperintensities and total vascular burden), and whether cerebrovascular lesions and brain amyloid load are associated with cognitive functioning. Methods: This exposure-to-control follow-up study examined 60 volunteers without dementia (mean age 70.9 years) with neurocognitive testing, brain 3T-MRI and amyloid-PET imaging. The volunteers were recruited from the Finnish Health 2000 survey (n = 6062) to attend follow-up examinations in 2014–2016 according to their insulin sensitivity in 2000 and their APOE genotype. The exposure group (n = 30) had IR in 2000 and the 30 controls had normal insulin sensitivity. There were 15 APOEɛ4 carriers per group. Statistical analyses were performed with multivariable linear models. Results: At follow-up the IR+group performed worse on executive functions (p = 0.02) and processing speed (p = 0.007) than the IR- group. The groups did not differ in cerebrovascular lesions. No associations were found between cerebrovascular lesions and neurocognitive test scores. Brain amyloid deposition associated with slower processing speed. Conclusion: Midlife IR predicted poorer executive functions and slower processing speed, but not cerebrovascular lesions. Brain amyloid deposition was associated with slower processing speed. The association between midlife IR and late-life cognition might not be mediated through cerebrovascular lesions measured here.
Collapse
Affiliation(s)
- Sini Toppala
- Turku PET Centre, University of Turku, Finland.,Turku City Hospital, University of Turku, Finland
| | | | | | - Semi Helin
- Turku PET Centre, University of Turku, Finland
| | - Saija Hurme
- Department of Biostatistics, University of Turku, Finland
| | - Jarkko Johansson
- Turku PET Centre, University of Turku, Finland.,Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | - Antti Jula
- National Institute for Health and Welfare, Turku, Finland
| | - Mira Karrasch
- Department of Psychology, Åbo Akademi University, Turku, Finland
| | | | - Hanna Laine
- Turku City Hospital, University of Turku, Finland.,Department of Medicine, University of Turku, Turku University Hospital, Turku, Finland
| | - Riitta Parkkola
- Department of Radiology, Turku University and Turku University Hospital, Turku, Finland
| | - Matti Viitanen
- Turku City Hospital, University of Turku, Finland.,Clinical Geriatrics, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | - Juha O Rinne
- Turku PET Centre, University of Turku, Finland.,Division of Clinical Neurosciences, Turku University Hospital, Turku, Finland
| |
Collapse
|
31
|
Mehra A, Sangwan G, Grover S, Kathirvel S, Avasthi A. Prevalence of Psychiatric Morbidity and Cognitive Impairment among Patients Attending the Rural Noncommunicable Disease Clinic. J Neurosci Rural Pract 2020; 11:585-592. [PMID: 33144795 PMCID: PMC7595800 DOI: 10.1055/s-0040-1715540] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Objective
This study aimed to assess the prevalence of cognitive impairment and psychiatric morbidity among the patients attending the rural noncommunicable disease clinic after controlling for various confounders (i.e., psychological morbidity, obesity, gender, level of education, duration of the illness and age).
Materials and Methods
One-hundred twenty-four patients were evaluated on the Hindi Mental State Examination for the cognitive function, Physical Health Questionnaire-9 for depression, and Generalized Anxiety Disorder-7 for anxiety disorders.
Results
About one-fourth (26.6%) of the participants had cognitive impairment. The prevalence of cognitive impairment was more among patients with hypertension (35.5%) as compared with the diabetes mellitus (13.6%) and those with comorbid hypertension and diabetes mellitus (26.6%). About one of the participants had depression (35.5%) and 29% of the patients had anxiety disorder. No significant difference was found in the level of cognitive deficits between those with hypertension and diabetes mellitus, when the confounding factors were not taken into account in the analysis. However, after controlling for psychiatric morbidity, obesity, gender, level of education, duration of the illness and age, those with hypertension were found to have significantly higher level of cognitive impairment compared with those with diabetes mellitus. A higher level of dysfunction was seen in the domains of orientation, registration, attention, recall, language, and visuospatial domains.
Conclusion
Present study suggests that patients of hypertension have higher level of cognitive impairment, when compared with those with diabetes mellitus, even after controlling for various confounders. Lack of difference between the two groups can be accounted by the confounding variables.
Collapse
Affiliation(s)
- Aseem Mehra
- Department of Psychiatry, Postgraduate Institute Medical Education and Research, Chandigarh, Punjab, India
| | - Garima Sangwan
- Department of Community Medicine and School of Public Health, Postgraduate Institute Medical Education and Research, Chandigarh, Punjab, India
| | - Sandeep Grover
- Department of Psychiatry, Postgraduate Institute Medical Education and Research, Chandigarh, Punjab, India
| | - Soundappan Kathirvel
- Department of Community Medicine and School of Public Health, Postgraduate Institute Medical Education and Research, Chandigarh, Punjab, India
| | - Ajit Avasthi
- Department of Psychiatry, Postgraduate Institute Medical Education and Research, Chandigarh, Punjab, India
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW Research has consistently shown that type 2 diabetes (T2D) is associated with increased risk of all-cause dementia. Because one of the most common clinical presentations of early stage dementia is memory impairment, we examined the relationship of T2D with memory function, using the recently published scientific literature. RECENT FINDINGS We conducted a structured review to identify studies of "T2D and memory" published since 2015. After review of the 129 articles retrieved, we identified 14 studies meeting the inclusion and exclusion criteria. Among the eight studies with a single assessment of memory function in time (mostly cross-sectional), six found an association of T2D with lower memory function, but mostly in select subgroups of persons. Separately, six studies included repeated measures of memory (longitudinal design). Four out of six longitudinal studies found that T2D was related with a faster decline in memory, while two did not. Among the four studies showing a relation with memory decline, two had sample sizes of 9000-10,000 persons. Further, three longitudinal studies controlled for hypertension and stroke as covariates, and results suggested that common vascular risk factors and diseases do not account for the relation. While mechanistic studies clearly support a role for cerebrovascular disease in the relation of T2D with cognition, emerging data suggest that insulin resistance in the brain itself may also play a role. Most, but not all, recently published studies suggest that T2D is associated with a lower level and faster decline in memory function. This association does not appear to be fully accounted for by common vascular processes. More research will clarify the mechanisms linking T2D to memory and dementia.
Collapse
|
33
|
Peeters G, Almirall Sanchez A, Llibre Guerra J, Lawlor B, Kenny RA, Yaffe K, Llibre Rodriguez J. Risk Factors for Incident Dementia Among Older Cubans. Front Public Health 2020; 8:481. [PMID: 33014976 PMCID: PMC7511701 DOI: 10.3389/fpubh.2020.00481] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/28/2020] [Indexed: 12/16/2022] Open
Abstract
Introduction: Little is known about risk factors of dementia in Latin American countries. We aimed to identify socio–demographic, health and lifestyle risk factors of incident dementia in Cuban older adults. Methods: Data were from 1,846 participants in the Cuban cohort of the 10/66 Dementia Research Group. Participants completed questionnaires, health examinations, and cognitive tests at baseline (2003–2006) and 4.5 years later (2007–2010). Associations between risk factors (baseline) and incident dementia (follow-up) were examined using logistic regression. Results: Just over 9% of participants developed dementia. Overall, older age and low physical activity were associated with incident dementia. In those 65–74 years of age, depression, stroke and low physical activity were associated with incident dementia. In those ≥75 years of age, low physical activity, never eating fish, and smoking were associated with incident dementia. Conclusions: Modifiable lifestyle factors play an important role in developing dementia in Cuban older adults. This knowledge opens up opportunities for preventive strategies.
Collapse
Affiliation(s)
- Geeske Peeters
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland.,Global Brain Health Institute, University of California San Francisco, San Francisco, CA, United States
| | - Arianna Almirall Sanchez
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland.,Global Brain Health Institute, University of California San Francisco, San Francisco, CA, United States
| | - Jorge Llibre Guerra
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland.,Global Brain Health Institute, University of California San Francisco, San Francisco, CA, United States.,Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
| | - Brian Lawlor
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland.,Global Brain Health Institute, University of California San Francisco, San Francisco, CA, United States.,Department of Psychiatry, Mercer's Institute for Successful Ageing, St. James's Hospital, Dublin, Ireland
| | - Rose Anne Kenny
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland.,Global Brain Health Institute, University of California San Francisco, San Francisco, CA, United States.,The Irish Longitudinal Study on Ageing, Trinity College Dublin, Dublin, Ireland.,The Irish Longitudinal Study on Ageing, Trinity College Dublin, Dublin, Ireland
| | - Kristine Yaffe
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland.,Global Brain Health Institute, University of California San Francisco, San Francisco, CA, United States.,Department of Psychiatry, Neurology and Epidemiology, University of California, San Francisco, San Francisco, CA, United States
| | - Juan Llibre Rodriguez
- Facultad de Medicina Finley-Albarrán, Universidad de Ciencias Médicas de la Habana, Havana, Cuba
| |
Collapse
|
34
|
Kellar D, Craft S. Brain insulin resistance in Alzheimer's disease and related disorders: mechanisms and therapeutic approaches. Lancet Neurol 2020; 19:758-766. [DOI: 10.1016/s1474-4422(20)30231-3] [Citation(s) in RCA: 201] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 05/30/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022]
|
35
|
Affleck AJ, Sachdev PS, Stevens J, Halliday GM. Antihypertensive medications ameliorate Alzheimer's disease pathology by slowing its propagation. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2020; 6:e12060. [PMID: 32802934 PMCID: PMC7424255 DOI: 10.1002/trc2.12060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/19/2020] [Accepted: 07/09/2020] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Mounting evidence supports an association between antihypertensive medication use and reduced risk of Alzheimer's disease (AD). Consensus on possible pathological mechanisms remains elusive. METHODS Human brain tissue from a cohort followed to autopsy that included 96 cases of AD (46 medicated for hypertension) and 53 pathological controls (33 also medicated) matched for cerebrovascular disease was available from the New South Wales Brain Banks. Quantified frontal cortex amyloid beta (Aβ) and tau proteins plus Alzheimer's neuropathologic change scores were analyzed. RESULTS Univariate analyses found no difference in amounts of AD proteins in the frontal cortex between medication users, but multivariate analyses showed that antihypertensive medication use was associated with a less extensive spread of AD proteins throughout the brain. DISCUSSION The heterogeneous nature of the antihypertensive medications is consistent with downstream beneficial effects of blood pressure lowering and/or management being associated with the reduced spreading of AD pathology observed.
Collapse
Affiliation(s)
- Andrew J. Affleck
- Neuroscience Research Australia (NeuRA)SydneyAustralia
- School of PsychiatryUniversity of New South WalesSydneyAustralia
| | - Perminder S. Sachdev
- Centre for Healthy Brain Ageing (CHeBA)School of Psychiatry Faculty of MedicineUniversity of New South WalesSydneyAustralia
| | - Julia Stevens
- Discipline of PathologySchool of Medical SciencesUniversity of SydneySydneyAustralia
| | - Glenda M. Halliday
- Neuroscience Research Australia (NeuRA)SydneyAustralia
- School of Medical SciencesFaculty of MedicineUniversity of New South WalesSydneyAustralia
- Brain and Mind Centre & Faculty of Medicine and HealthSydney Medical SchoolUniversity of SydneySydneyAustralia
| |
Collapse
|
36
|
Biessels GJ, Nobili F, Teunissen CE, Simó R, Scheltens P. Understanding multifactorial brain changes in type 2 diabetes: a biomarker perspective. Lancet Neurol 2020; 19:699-710. [PMID: 32445622 DOI: 10.1016/s1474-4422(20)30139-3] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 03/20/2020] [Accepted: 04/08/2020] [Indexed: 12/14/2022]
Abstract
People with type 2 diabetes are at an increased risk of cognitive impairment and dementia (including Alzheimer's disease), as well as subtle forms of cognitive dysfunction. Current diabetes guidelines recommend screening for cognitive impairment in groups at high risk and providing guidance for diabetes management in patients with diabetes and cognitive impairment. Yet, no disease-modifying treatment is available and important questions remain about the mechanisms underlying diabetes-associated cognitive dysfunction. These mechanisms are likely to be multifactorial and different for subtle and more severe forms of diabetes-associated cognitive dysfunction. Over the past years, research on dementia, brain ageing, diabetes, and vascular disease has identified novel biomarkers of specific dementia aetiologies, brain parenchymal injury, and cerebral blood flow and metabolism. These markers shed light on the processes underlying diabetes-associated cognitive dysfunction, have clear applications in current research and increasingly in clinical diagnosis, and might ultimately guide targeted treatment.
Collapse
Affiliation(s)
- Geert Jan Biessels
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, Netherlands.
| | - Flavio Nobili
- Department of Neuroscience, Ophthalmology, Genetics, and Child and Mother Health, University of Genoa, Genoa, Italy; Clinical Neurology Unit, IRCSS Ospedale Policlinico San Martino, Genoa, Italy
| | - Charlotte E Teunissen
- Neurochemistry Laboratory and Biobank, Department of Clinical Chemistry, Amsterdam, Netherlands
| | - Rafael Simó
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | - Philip Scheltens
- Department of Neurology and Alzheimer Center, VU University Medical Center Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
37
|
Kubis-Kubiak A, Dyba A, Piwowar A. The Interplay between Diabetes and Alzheimer's Disease-In the Hunt for Biomarkers. Int J Mol Sci 2020; 21:ijms21082744. [PMID: 32326589 PMCID: PMC7215807 DOI: 10.3390/ijms21082744] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/09/2020] [Accepted: 04/12/2020] [Indexed: 02/07/2023] Open
Abstract
The brain is an organ in which energy metabolism occurs most intensively and glucose is an essential and dominant energy substrate. There have been many studies in recent years suggesting a close relationship between type 2 diabetes mellitus (T2DM) and Alzheimer’s disease (AD) as they have many pathophysiological features in common. The condition of hyperglycemia exposes brain cells to the detrimental effects of glucose, increasing protein glycation and is the cause of different non-psychiatric complications. Numerous observational studies show that not only hyperglycemia but also blood glucose levels near lower fasting limits (72 to 99 mg/dL) increase the incidence of AD, regardless of whether T2DM will develop in the future. As the comorbidity of these diseases and earlier development of AD in T2DM sufferers exist, new AD biomarkers are being sought for etiopathogenetic changes associated with early neurodegenerative processes as a result of carbohydrate disorders. The S100B protein seem to be interesting in this respect as it may be a potential candidate, especially important in early diagnostics of these diseases, given that it plays a role in both carbohydrate metabolism disorders and neurodegenerative processes. It is therefore necessary to clarify the relationship between the concentration of the S100B protein and glucose and insulin levels. This paper draws attention to a valuable research objective that may in the future contribute to a better diagnosis of early neurodegenerative changes, in particular in subjects with T2DM and may be a good basis for planning experiments related to this issue as well as a more detailed explanation of the relationship between the neuropathological disturbances and changes of glucose and insulin concentrations in the brain.
Collapse
Affiliation(s)
- Adriana Kubis-Kubiak
- Department of Toxicology, Faculty of Pharmacy, Wroclaw Medical University, 50367 Wroclaw, Poland;
- Correspondence:
| | - Aleksandra Dyba
- Students Science Club of the Department of Toxicology, Faculty of Pharmacy, Wroclaw Medical University, 50367 Wroclaw, Poland;
| | - Agnieszka Piwowar
- Department of Toxicology, Faculty of Pharmacy, Wroclaw Medical University, 50367 Wroclaw, Poland;
| |
Collapse
|
38
|
Shinohara M, Tashiro Y, Suzuki K, Fukumori A, Bu G, Sato N. Interaction between APOE genotype and diabetes in cognitive decline. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2020; 12:e12006. [PMID: 32211501 PMCID: PMC7085280 DOI: 10.1002/dad2.12006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/10/2019] [Accepted: 11/01/2019] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Although diabetes and apolipoprotein E (apoE) are both significant risk factors for dementia, including Alzheimer's disease, it remains to be clarified how they are related to each other in contributing to the risk of dementia. METHODS By reviewing the National Alzheimer's Coordinating Center (NACC) clinical records, we investigated whether diabetes affects cognitive decline depending on APOE genotype and their potential relationships with neuropathology. RESULTS A significant interaction between diabetes and APOE genotype exists, where diabetes affected cognitive decline in APOE3 carriers and APOE2 carriers, but not APOE4 carriers. Moreover, the presence of vascular pathology was increased by diabetes in APOE3 carriers, while APOE4 carriers nearly reached plateau levels irrespective of diabetes. DISCUSSION Diabetes accelerates cognitive decline, in part, through accelerating vascular impairment in non-APOE ε4 carriers, but such effects are negligible in APOE4 carriers, who themselves are already vulnerable to vascular impairment.
Collapse
Affiliation(s)
- Mitsuru Shinohara
- Department of Aging NeurobiologyCenter for Development of Advanced Medicine for DementiaNational Center for Geriatrics and GerontologyObuAichiJapan
- Department of Aging NeurobiologyGraduate School of MedicineOsaka UniversitySuitaOsakaJapan
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Yoshitaka Tashiro
- Department of Aging NeurobiologyCenter for Development of Advanced Medicine for DementiaNational Center for Geriatrics and GerontologyObuAichiJapan
| | - Kaoru Suzuki
- Department of Aging NeurobiologyCenter for Development of Advanced Medicine for DementiaNational Center for Geriatrics and GerontologyObuAichiJapan
| | - Akio Fukumori
- Department of Aging NeurobiologyCenter for Development of Advanced Medicine for DementiaNational Center for Geriatrics and GerontologyObuAichiJapan
- Department of Aging NeurobiologyGraduate School of MedicineOsaka UniversitySuitaOsakaJapan
| | - Guojun Bu
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Naoyuki Sato
- Department of Aging NeurobiologyCenter for Development of Advanced Medicine for DementiaNational Center for Geriatrics and GerontologyObuAichiJapan
- Department of Aging NeurobiologyGraduate School of MedicineOsaka UniversitySuitaOsakaJapan
| |
Collapse
|
39
|
Sanchis-Soler G, Tortosa-Martínez J, Manchado-Lopez C, Cortell-Tormo JM. The effects of stress on cardiovascular disease and Alzheimer's disease: Physical exercise as a counteract measure. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 152:157-193. [PMID: 32450995 DOI: 10.1016/bs.irn.2020.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
AD is a complicated multi-systemic neurological disorder that involves different biological pathways. Several risk factors have been identified, including chronic stress. Chronic stress produces an alteration in the activity of the hypothalamic pituitary adrenal (HPA) system, and the autonomic nervous system (ANS), which over time increase the risk of AD and also the incidence of cardiovascular disease (CVD) and risk factors, such as hypertension, obesity and type 2 diabetes, associated with cognitive impairment and AD. Considering the multi-factorial etiology of AD, understanding the complex interrelationships between different risk factors is of potential interest for designing adequate strategies for preventing, delaying the onset or slowing down the progression of this devastating disease. Thus, in this review we will explore the general mechanisms and evidence linking stress, cardiovascular disease and AD, and discuss the potential benefits of physical activity for AD by counteracting the negative effects of chronic stress, CVD and risk factors.
Collapse
|
40
|
Luchsinger JA, Palta P, Rippon B, Sherwood G, Soto L, Ceballos F, Laing K, Igwe K, Tomljanovic Z, He H, Razlighi Q, Teresi J, Moreno H, Brickman AM. Pre-Diabetes, but not Type 2 Diabetes, Is Related to Brain Amyloid in Late Middle-Age. J Alzheimers Dis 2020; 75:1241-1252. [PMID: 32390636 PMCID: PMC7659021 DOI: 10.3233/jad-200232] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Type 2 diabetes is a dementia risk factor, but its relation to Alzheimer's disease (AD), the most common cause of dementia, is unclear. OBJECTIVE Our primary objective was to examine the association of pre-diabetes and type 2 diabetes with brain amyloid-β (Aβ), the putative main culprit of AD. Our secondary objective was to examine the association of pre-diabetes and type 2 diabetes with neurodegeneration, cerebrovascular disease (CVD), and memory performance. METHODS We conducted a cross-sectional study of 350 late middle-aged Hispanics without dementia in New York City. We classified diabetes status as normal glucose tolerance (NGT), pre-diabetes, and type 2 diabetes following American Diabetes Association criteria. Brain Aβ was ascertained as global Aβ standardized value uptake ratio using PET with 18F-Florbetaben. Neurodegeneration was operationalized as cortical thickness in regions affected by AD using MRI. CVD was operationalized as white matter hyperintensity volume (WMH) on MRI, and memory as performance with the selective reminding test (SRT). RESULTS Mean age was 64.15±3.34 years, 72.00% were women, and 35.43% were APOEɛ4 carriers. Pre-diabetes, but not type 2 diabetes, was associated with higher Aβ compared with NGT. Type 2 diabetes treatment was related to lower Aβ. Type 2 diabetes was related to lower cortical thickness, higher WMH, and lower SRT score. CONCLUSION Pre-diabetes, but not type 2 diabetes, is associated with higher brain Aβ in late middle age, and this observation could be explained by the relation of diabetes treatment with lower brain Aβ. Whether type 2 diabetes treatment lowers brain Aβ requires further study.
Collapse
Affiliation(s)
- Jose A. Luchsinger
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center (CUIMC), New York, NY
- Department of Epidemiology, Joseph P. Mailman School of Public Health, CUIMC, New York, NY
| | - Priya Palta
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center (CUIMC), New York, NY
- Department of Epidemiology, Joseph P. Mailman School of Public Health, CUIMC, New York, NY
| | - Brady Rippon
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center (CUIMC), New York, NY
| | - Greysi Sherwood
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center (CUIMC), New York, NY
| | - Luisa Soto
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center (CUIMC), New York, NY
| | - Fernando Ceballos
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center (CUIMC), New York, NY
| | - Krystal Laing
- Department of Neurology, College of Physicians and Surgeons, CUIMC, New York, NY
| | - Kay Igwe
- Department of Neurology, College of Physicians and Surgeons, CUIMC, New York, NY
| | - Zeljko Tomljanovic
- Department of Neurology, College of Physicians and Surgeons, CUIMC, New York, NY
| | - Hengda He
- Department of Neurology, College of Physicians and Surgeons, CUIMC, New York, NY
| | - Qolamreza Razlighi
- Department of Neurology, College of Physicians and Surgeons, CUIMC, New York, NY
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, CUIMC, New York, NY
- Gertrude H. Sergievsky Center, CUIMC, New York, NY
- Department of Biomedical Engineering, Columbia University, New York, NY
| | - Jeanne Teresi
- Research Division, Hebrew Home in Riverdale, Bronx, NY
| | - Herman Moreno
- Department of Neurology, SUNY Downstate Medical Center, Brooklyn, NY
| | - Adam M. Brickman
- Department of Neurology, College of Physicians and Surgeons, CUIMC, New York, NY
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, CUIMC, New York, NY
- Gertrude H. Sergievsky Center, CUIMC, New York, NY
| |
Collapse
|
41
|
Conner SC, Pase MP, Carneiro H, Raman MR, McKee AC, Alvarez VE, Walker JM, Satizabal CL, Himali JJ, Stein TD, Beiser A, Seshadri S. Mid-life and late-life vascular risk factor burden and neuropathology in old age. Ann Clin Transl Neurol 2019; 6:2403-2412. [PMID: 31691546 PMCID: PMC6917310 DOI: 10.1002/acn3.50936] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE To determine whether vascular risk factor burden in mid- or late-life associates with postmortem vascular and neurodegenerative pathologies in a community-based sample. METHODS We studied participants from the Framingham Heart Study who participated in our voluntary brain bank program. Overall vascular risk factor burden was calculated using the Framingham Stroke Risk Profile (FSRP). Mid-life FSRP was measured at 50 to 60 years of age. Following death, brains were autopsied and semi-quantitatively assessed by board-certified neuropathologists for cerebrovascular outcomes (cortical infarcts, subcortical infarcts, atherosclerosis, arteriosclerosis) and Alzheimer's disease pathology (Braak stage, cerebral amyloid angiopathy, and neuritic plaque score). We estimated adjusted odds ratios between vascular risk burden (at mid-life and before death) and neuropathological outcomes using logistic and proportional-odds logistic models. RESULTS The median time interval between FSRP and death was 33.4 years for mid-life FSRP and 4.4 years for final FSRP measurement before death. Higher mid-life vascular risk burden was associated with increased odds of all cerebrovascular pathology, even with adjustment for vascular risk burden before death. Late-life vascular risk burden was associated with increased odds of cortical infarcts (OR [95% CI]: 1.04 [1.00, 1.08]) and arteriosclerosis stage (OR [95% CI]: 1.03 [1.00, 1.05]). Mid-life vascular risk burden was not associated with Alzheimer's disease pathology, though late-life vascular risk burden was associated with increased odds of higher Braak stage (OR [95% CI]: 1.03 [1.01, 1.05]). INTERPRETATION Mid-life vascular risk burden was predictive of cerebrovascular but not Alzheimer's disease neuropathology, even after adjustment for vascular risk factors before death.
Collapse
Affiliation(s)
- Sarah C. Conner
- Framingham Heart StudyFraminghamMassachusetts
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusetts
| | - Matthew P. Pase
- Framingham Heart StudyFraminghamMassachusetts
- Melbourne Dementia Research CentreThe Florey Institute for Neuroscience and Mental HealthMelbourneAustralia
- Centre for Human PsychopharmacologySwinburne University of TechnologyMelbourneAustralia
- Faculty of MedicineDentistry and Health SciencesThe University of MelbourneMelbourneAustralia
| | - Herman Carneiro
- Framingham Heart StudyFraminghamMassachusetts
- Department of MedicineBoston University School of MedicineBostonMassachusetts
| | - Mekala R. Raman
- Department of NeurologyBoston University School of MedicineBostonMassachusetts
| | - Ann C. McKee
- Department of NeurologyBoston University School of MedicineBostonMassachusetts
- Boston UniversityAlzheimer's Disease and CTE CenterBoston University School of MedicineBostonMassachusetts
- Department of Veterans Affairs Medical CenterBedfordMassachusetts
- VA Boston Healthcare SystemBostonMassachusetts
- Department of Pathology and Laboratory MedicineBoston University School of MedicineBostonMassachusetts
| | - Victor E. Alvarez
- Boston UniversityAlzheimer's Disease and CTE CenterBoston University School of MedicineBostonMassachusetts
- Department of Veterans Affairs Medical CenterBedfordMassachusetts
- VA Boston Healthcare SystemBostonMassachusetts
- Department of Pathology and Laboratory MedicineBoston University School of MedicineBostonMassachusetts
| | - Jamie M. Walker
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative DiseasesUT Health San AntonioSan AntonioTexas
| | - Claudia L. Satizabal
- Framingham Heart StudyFraminghamMassachusetts
- Department of NeurologyBoston University School of MedicineBostonMassachusetts
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative DiseasesUT Health San AntonioSan AntonioTexas
| | - Jayandra J. Himali
- Framingham Heart StudyFraminghamMassachusetts
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusetts
- Department of NeurologyBoston University School of MedicineBostonMassachusetts
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative DiseasesUT Health San AntonioSan AntonioTexas
| | - Thor D. Stein
- Boston UniversityAlzheimer's Disease and CTE CenterBoston University School of MedicineBostonMassachusetts
- Department of Veterans Affairs Medical CenterBedfordMassachusetts
- VA Boston Healthcare SystemBostonMassachusetts
- Department of Pathology and Laboratory MedicineBoston University School of MedicineBostonMassachusetts
| | - Alexa Beiser
- Framingham Heart StudyFraminghamMassachusetts
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusetts
- Department of NeurologyBoston University School of MedicineBostonMassachusetts
| | - Sudha Seshadri
- Framingham Heart StudyFraminghamMassachusetts
- Department of NeurologyBoston University School of MedicineBostonMassachusetts
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative DiseasesUT Health San AntonioSan AntonioTexas
| |
Collapse
|
42
|
Nasoohi S, Parveen K, Ishrat T. Metabolic Syndrome, Brain Insulin Resistance, and Alzheimer's Disease: Thioredoxin Interacting Protein (TXNIP) and Inflammasome as Core Amplifiers. J Alzheimers Dis 2019; 66:857-885. [PMID: 30372683 DOI: 10.3233/jad-180735] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Empirical evidence indicates a strong association between insulin resistance and pathological alterations related to Alzheimer's disease (AD) in different cerebral regions. While cerebral insulin resistance is not essentially parallel with systemic metabolic derangements, type 2 diabetes mellitus (T2DM) has been established as a risk factor for AD. The circulating "toxic metabolites" emerging in metabolic syndrome may engage several biochemical pathways to promote oxidative stress and neuroinflammation leading to impair insulin function in the brain or "type 3 diabetes". Thioredoxin-interacting protein (TXNIP) as an intracellular amplifier of oxidative stress and inflammasome activation may presumably mediate central insulin resistance. Emerging data including those from our recent studies has demonstrated a sharp TXNIP upregulation in stroke, aging and AD and well underlining the significance of this hypothesis. With the main interest to illustrate TXNIP place in type 3 diabetes, the present review primarily briefs the potential mechanisms contributing to cerebral insulin resistance in a metabolically deranged environment. Then with a particular focus on plausible TXNIP functions to drive and associate with AD pathology, we present the most recent evidence supporting TXNIP as a promising therapeutic target in AD as an age-associated dementia.
Collapse
|
43
|
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
|
44
|
Diabetes drugs in the fight against Alzheimer's disease. Ageing Res Rev 2019; 54:100936. [PMID: 31330313 DOI: 10.1016/j.arr.2019.100936] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/20/2019] [Accepted: 07/17/2019] [Indexed: 01/10/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia, particularly in old age subjects. Hyperinsulinemia and insulin resistance, which are known as pathophysiological features of Type 2 Diabetes Mellitus (T2DM), have also been demonstrated to have a significant impact on cognitive impairment. Studies have shown that an altered insulin pathway may interact with amyloid-β protein deposition and tau protein phosphorylation, both leading factors for AD development. Drugs used for T2DM treatment from insulin and metformin through dipeptidyl peptidase-4 inhibitors and glucagon-like peptide-1 receptor agonists may represent a promising approach to fight AD. With this review from animal to human studies, we aim at responding to the reasons why drugs for diabetes may represent potential treatments for AD.
Collapse
|
45
|
Wanleenuwat P, Iwanowski P, Kozubski W. Alzheimer's dementia: pathogenesis and impact of cardiovascular risk factors on cognitive decline. Postgrad Med 2019; 131:415-422. [PMID: 31424301 DOI: 10.1080/00325481.2019.1657776] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia manifesting as alterations in cognitive abilities, behavior, and deterioration in memory which is progressive, leading to gradual worsening of symptoms. Major pathological features of AD are accumulations of neuronal amyloid plaques and neurofibrillary tangles, with early lesions appearing primarily in the hippocampus, the area of the brain involved in memory and learning. Cardiovascular-related risk factors are believed to play a crucial role in disease development and the acceleration of cognitive deterioration by worsening cerebral perfusion, promoting disturbances in amyloid clearance. Current evidence supports hypertension, hypotension, heart failure, stroke and coronary artery diseases as potential factors playing a role in cognitive decline in patients with Alzheimer's dementia. Although dementia due to cardiovascular deficits is more strongly linked to the development of vascular dementia, a stepwise decline in cognition, recent researches have also discovered its deleterious influence on AD development.
Collapse
Affiliation(s)
- Pitchaya Wanleenuwat
- Department of Neurology, Poznan University of Medical Sciences , Poznan , Poland
| | - Piotr Iwanowski
- Department of Neurology, Poznan University of Medical Sciences , Poznan , Poland
| | - Wojciech Kozubski
- Department of Neurology, Poznan University of Medical Sciences , Poznan , Poland
| |
Collapse
|
46
|
Stepler KE, Robinson RAS. The Potential of ‘Omics to Link Lipid Metabolism and Genetic and Comorbidity Risk Factors of Alzheimer’s Disease in African Americans. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1118:1-28. [DOI: 10.1007/978-3-030-05542-4_1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
47
|
Bello-Chavolla OY, Antonio-Villa NE, Vargas-Vázquez A, Ávila-Funes JA, Aguilar-Salinas CA. Pathophysiological Mechanisms Linking Type 2 Diabetes and Dementia: Review of Evidence from Clinical, Translational and Epidemiological Research. Curr Diabetes Rev 2019; 15:456-470. [PMID: 30648514 DOI: 10.2174/1573399815666190129155654] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/26/2018] [Accepted: 01/08/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Type 2 diabetes represents an increasing health burden world-wide and its prevalence in particularly higher in elderly population. Consistent epidemiological evidence suggests an increased risk of dementia associated to type 2 diabetes; the mechanisms underlying these associations, however, remain unclear. OBJECTIVE The study aims to review epidemiological, clinical and pre-clinical data that weigh on pathophysiological links, mechanisms of disease and associations between type 2 diabetes and dementia to identify areas of opportunity for future research. METHODS We searched the following electronic bibliographic databases: PUBMED, EMBASE, SCIELO, MEDLINE and OVID for clinical, translational and epidemiological research literature that summarize diabetes-related risk factors for dementia, metabolic and neurological changes associated to T2D, evidence of therapeutic approaches in type 2 diabetes and its pathophysiological implications for dementia. RESULTS Type 2 diabetes mellitus increases risk for all-cause dementia, vascular dementia and Alzheimer's disease. The most evaluated mechanisms linking both disorders in pre-clinical studies include an increase in neuronal insulin resistance, impaired insulin signaling, pro-inflammatory state, mitochondrial dysfunction and vascular damage which increase deposition of β-amyloid, tau proteins and GSK3β, leading to an earlier onset of dementia in individuals with impairment in the glucose metabolism. Neuroimaging and neuropathology evidence linking cerebrovascular lesions, neurodegeneration and particularly small-vessel disease in the onset of dementia is consistent with the increased risk of incident dementia in type 2 diabetes, but consistent evidence of AD-related pathology is scarce. Epidemiological data shows increased risk of dementia related to hypoglycemic episodes, glycemic control, metabolic syndrome, insulin resistance and genetic predisposition, but the evidence is not consistent and statistical analysis might be affected by inconsistent covariate controlling. Therapeutic approaches for T2D have shown inconsistent result in relation to dementia prevention and delay of cognitive decline; lifestyle intervention, particularly physical activity, is a promising alternative to ameliorate the impact of disability and frailty on T2D-related dementia. CONCLUSION Vascular disease, inflammation and impaired brain insulin signaling might occur in T2D and contribute to dementia risk. Evidence from epidemiological studies has not consistently reported associations that could integrate a unified mechanism of disease in humans. Evaluation of the effect of antidiabetic medications and non-pharmacological interventions in dementia prevention in type 2 diabetes is promising but has thus far offered inconsistent results.
Collapse
Affiliation(s)
- Omar Yaxmehen Bello-Chavolla
- Metabolic Disease Research Unit, National Institute of Medical Sciences and Nutrition, Mexico, United States
- Faculty of Medicine, National Autonomous, University of Mexico, Mexico, United States
| | - Neftali Eduardo Antonio-Villa
- Metabolic Disease Research Unit, National Institute of Medical Sciences and Nutrition, Mexico, United States
- Faculty of Medicine, National Autonomous, University of Mexico, Mexico, United States
| | - Arsenio Vargas-Vázquez
- Metabolic Disease Research Unit, National Institute of Medical Sciences and Nutrition, Mexico, United States
- Faculty of Medicine, National Autonomous, University of Mexico, Mexico, United States
| | - José Alberto Ávila-Funes
- Department of Geriatrics, National Institute of Medical Sciences and Nutrition, Mexico, United States
- University of Bordeaux, Bordeaux Population Health Research Center, 33076 Bordeaux, France
| | - Carlos Alberto Aguilar-Salinas
- Department of Endocrinology and Metabolism, National Institute of Medical Sciences and Nutrition, Mexico, United States
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Sertoma, 64710 Monterrey, N.L., Mexico, United States
| |
Collapse
|
48
|
Abstract
Population-based clinic-pathological studies have established that the most common pathological substrate of dementia in community-dwelling elderly people is mixed, especially Alzheimer's disease (AD) and cerebrovascular ischemic disease (CVID), rather than pure AD. While these could be just two frequent unrelated comorbidities in the elderly, epidemiological research has reinforced the idea that mid-life (age <65 years) vascular risk factors increase the risk of late-onset (age ≥ 65 years) dementia, and specifically AD. By contrast, healthy lifestyle choices such as leisure activities, physical exercise, and Mediterranean diet are considered protective against AD. Remarkably, several large population-based longitudinal epidemiological studies have recently indicated that the incidence and prevalence of dementia might be decreasing in Western countries. Although it remains unclear whether these positive trends are attributable to neuropathologically definite AD versus CVID, based on these epidemiological data it has been estimated that a sizable proportion of AD cases could be preventable. In this review, we discuss the current evidence about modifiable risk factors for AD derived from epidemiological, preclinical, and interventional studies, and analyze the opportunities for therapeutic and preventative interventions.
Collapse
Affiliation(s)
- Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - John H. Growdon
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
49
|
Groeneveld O, Reijmer Y, Heinen R, Kuijf H, Koekkoek P, Janssen J, Rutten G, Kappelle L, Biessels G. Brain imaging correlates of mild cognitive impairment and early dementia in patients with type 2 diabetes mellitus. Nutr Metab Cardiovasc Dis 2018; 28:1253-1260. [PMID: 30355471 DOI: 10.1016/j.numecd.2018.07.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/07/2018] [Accepted: 07/24/2018] [Indexed: 11/21/2022]
Abstract
BACKGROUND AND AIMS The risk of mild cognitive impairment and dementia is increased in type 2 diabetes mellitus (T2DM). We aimed to identify the neuroanatomical correlates of mild cognitive impairment (MCI) and early dementia in patients with T2DM, using advanced multimodal MRI. METHODS AND RESULTS Twenty-five patients (≥70 years) with T2DM and MCI (n = 22) or early dementia (n = 3) were included. The reference group consisted of 23 patients with T2DM with intact cognition. All patients underwent a 3 T MRI. Brain volumes and white matter hyperintensity volumes were obtained with automated segmentation methods. White matter connectivity was assessed with diffusion tensor imaging and fiber tractography. Infarcts and microbleeds were rated visually. Compared to patients without cognitive impairment, those with impairment had a lower grey matter volume (effect size: -0.58, p=0.042), especially in the right temporal lobe and subcortical brain regions (effect sizes: -0.45 to -0.91, false discovery rate corrected p < 0.05). White matter volume (effect size: -0.47, p = 0.11) and white matter connectivity (effect size: 0.55, p = 0.054) were also reduced in patients with versus without cognitive impairment, albeit not statistically significant. White matter hyperintensity volumes and occurrence of other vascular lesions did not differ between the two patient groups. CONCLUSION In patients with T2DM, grey matter atrophy rather than vascular brain injury appears to be the primary imaging correlate of MCI and early dementia.
Collapse
Affiliation(s)
- O Groeneveld
- Brain Center Rudolf Magnus, Department of Neurology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands.
| | - Y Reijmer
- Brain Center Rudolf Magnus, Department of Neurology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - R Heinen
- Brain Center Rudolf Magnus, Department of Neurology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - H Kuijf
- Image Sciences Institute, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - P Koekkoek
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - J Janssen
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - G Rutten
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - L Kappelle
- Brain Center Rudolf Magnus, Department of Neurology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - G Biessels
- Brain Center Rudolf Magnus, Department of Neurology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| |
Collapse
|
50
|
Abstract
Cognitive dysfunction is increasingly recognized as an important comorbidity of diabetes mellitus. Different stages of diabetes-associated cognitive dysfunction exist, each with different cognitive features, affected age groups and prognoses and probably with different underlying mechanisms. Relatively subtle, slowly progressive cognitive decrements occur in all age groups. More severe stages, particularly mild cognitive impairment and dementia, with progressive deficits, occur primarily in older individuals (>65 years of age). Patients in the latter group are the most relevant for patient management and are the focus of this Review. Here, we review the evolving insights from studies on risk factors, brain imaging and neuropathology, which provide important clues on mechanisms of both the subtle cognitive decrements and the more severe stages of cognitive dysfunction. In the majority of patients, the cognitive phenotype is probably defined by multiple aetiologies. Although both the risk of clinically diagnosed Alzheimer disease and that of vascular dementia is increased in association with diabetes, the cerebral burden of the prototypical pathologies of Alzheimer disease (such as neurofibrillary tangles and neuritic plaques) is not. A major challenge for researchers is to pinpoint from the spectrum of diabetes-related disease processes those that affect the brain and contribute to development of dementia beyond the pathologies of Alzheimer disease. Observations from experimental models can help to meet that challenge, but this requires further improving the synergy between experimental and clinical scientists. The development of targeted treatment and preventive strategies will therefore depend on these translational efforts.
Collapse
Affiliation(s)
- Geert Jan Biessels
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands.
| | - Florin Despa
- Department of Pharmacology and Nutritional Sciences and Department of Neurology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|