101
|
Besser LM, Rodriguez DA, McDonald N, Kukull WA, Fitzpatrick AL, Rapp SR, Seeman T. Neighborhood built environment and cognition in non-demented older adults: The Multi-Ethnic Study of Atherosclerosis. Soc Sci Med 2018; 200:27-35. [PMID: 29355828 PMCID: PMC5893410 DOI: 10.1016/j.socscimed.2018.01.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 01/05/2018] [Accepted: 01/08/2018] [Indexed: 12/16/2022]
Abstract
Preliminary studies suggest that neighborhood social and built environment (BE) characteristics may affect cognition in older adults. Older adults are particularly vulnerable to the neighborhood environment due to a decreasing range of routine travel with increasing age. We examined if multiple neighborhood BE characteristics are cross-sectionally associated with cognition in a diverse sample of older adults, and if the BE-cognition associations vary by individual-level demographics. The sample included 4539 participants from the Multi-Ethnic Study of Atherosclerosis. Multivariable linear regression was used to examine the associations between five BE measures and four cognitive measures, and effect modification by individual-level education and race/ethnicity. In the overall sample, increasing social destination density, walking destination density, and intersection density were associated with worse overall cognition, whereas increasing proportion of land dedicated to retail was associated with better processing speed. Effect modification results suggest that the association between urban density and worse cognition may be limited to or strongest in those of non-white race/ethnicity. Although an increase in neighborhood retail destinations was associated with better cognition in the overall sample, these results suggest that certain BE characteristics in dense urban environments may have a disproportionately negative association with cognition in vulnerable populations. However, our findings must be replicated in longitudinal studies and other regional samples.
Collapse
Affiliation(s)
- Lilah M Besser
- National Alzheimer's Coordinating Center, Department of Epidemiology, University of Washington, 4311 11th Ave NE, Suite 300, Seattle, WA 98105, USA.
| | - Daniel A Rodriguez
- Department of City and Regional Planning, Institute for Transportation Studies, University of California, Berkeley, Office 313B, Wurster Hall #1820, Berkeley, CA, 94720, USA.
| | - Noreen McDonald
- Department of City and Regional Planning, University of North Carolina at Chapel Hill, 317 New East Building, CB 3140, Chapel Hill, NC 27599, USA.
| | - Walter A Kukull
- National Alzheimer's Coordinating Center, Department of Epidemiology, University of Washington, 4311 11th Ave NE, Suite 300, Seattle, WA 98105, USA.
| | - Annette L Fitzpatrick
- Departments of Family Medicine, Epidemiology and Global Health, University of Washington, 1959 NE Pacific Street, Health Sciences Building, F-350, Box 357230, Seattle, WA 98195, USA.
| | - Stephen R Rapp
- Department of Psychiatry and Behavioral Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC 27157, USA.
| | - Teresa Seeman
- Department of Medicine, Division of Geriatrics, University of California, Los Angeles, 10945 Le Conte Avenue, Suite 2339, Los Angeles, CA 90095, USA.
| |
Collapse
|
102
|
Sesti G, Antonelli Incalzi R, Bonora E, Consoli A, Giaccari A, Maggi S, Paolisso G, Purrello F, Vendemiale G, Ferrara N. Management of diabetes in older adults. Nutr Metab Cardiovasc Dis 2018; 28:206-218. [PMID: 29337017 DOI: 10.1016/j.numecd.2017.11.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/22/2017] [Accepted: 11/28/2017] [Indexed: 02/08/2023]
Abstract
Type 2 diabetes prevalence is high in older adults and is expected to rise in the next decades. Diabetes in the population of frail older adults is accompanied by functional disability, several comorbidities, and premature mortality. A comprehensive geriatric assessment, including functional, cognitive, mental and social status, is advisable for identifying the glycemic targets and glucose-lowering therapies, focused on patient preferences, needs, and risks. The therapeutic options for older adults with diabetes are like those for the adult population. However, the pharmacological treatments must be carefully prescribed and monitored, taking into consideration the patient cognitive capacities, the potentially life-threatening drug-drug interactions, the cardiovascular risk, and with the main goal of avoiding hypoglycemia. Also, a careful nutritional evaluation with appropriate tools, as well as a balanced and periodically monitored physical activity, contribute to an effective tailored care plan, as needed by older adults with diabetes. This review evaluates the currently available hypoglycemic drugs and the current indications to the Italian diabetology community, specifically with regard to the treatment of adults aged 75 years or older with diabetes, including the unmet needs by the guidelines.
Collapse
Affiliation(s)
- G Sesti
- University Magna Graecia of Catanzaro, Catanzaro, Italy.
| | - R Antonelli Incalzi
- Gerontology Unit, Campus Bio Medico University and Teaching Hospital, Rome, Italy
| | - E Bonora
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Verona University and Hospital, Verona, Italy
| | - A Consoli
- Department of Medicine and Ageing Sciences and Inter-Departmental Center CeSI-MeT, University d'Annunzio, Chieti, Italy
| | - A Giaccari
- Center for Endocrine and Metabolic Diseases, Fondazione Policlinico Universitario A. Gemelli, UCSC, Rome Italy
| | - S Maggi
- National Research Council, Neuroscience Institute-Aging Branch, Padova, Italy
| | - G Paolisso
- Università degli Studi della Campania - Luigi Vanvitelli, Italy
| | - F Purrello
- Department of Clinical and Experimental Medicine, University of, Catania, Italy
| | - G Vendemiale
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - N Ferrara
- University of Naples Federico II, Napoli, Italy
| |
Collapse
|
103
|
Bangen KJ, Himali JJ, Beiser AS, Nation DA, Libon DJ, Fox CS, Seshadri S, Wolf PA, McKee AC, Au R, Delano-Wood L. Interaction Between Midlife Blood Glucose and APOE Genotype Predicts Later Alzheimer's Disease Pathology. J Alzheimers Dis 2018; 53:1553-62. [PMID: 27392855 DOI: 10.3233/jad-160163] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Elevated blood glucose and the apolipoprotein (APOE) ɛ4 allele have both been associated with increased dementia risk; however, the neuropathological mechanisms underlying these associations remain unclear. We examined the impact of APOE genotype and midlife blood glucose on post-mortem vascular and Alzheimer's disease (AD) neuropathology. Ninety-four participants from the Framingham Heart Study without diagnosed diabetes underwent health examination at midlife and brain autopsy at death. Histopathological measures of vascular and AD neuropathology were obtained and analyzed. Results demonstrated that, among APOE ɛ4 carriers, elevated blood glucose was associated with more severe AD pathology. There was no such relationship with vascular pathology. In a relatively healthy sample with low vascular risk burden, midlife elevated blood glucose was associated with greater AD pathology among APOE ɛ4 carriers. A better understanding of interactive effects of APOE genotype and vascular risk on neuropathology has implications for identification of individuals at risk for decline and long-term preventive treatment.
Collapse
Affiliation(s)
- Katherine J Bangen
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Psychiatry, University of California, San Diego School of Medicine, La Jolla, CA, USA
| | - Jayandra J Himali
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,The Framingham Heart Study, Framingham, MA, USA
| | - Alexa S Beiser
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,The Framingham Heart Study, Framingham, MA, USA.,Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Daniel A Nation
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - David J Libon
- Department of Geriatrics and Gerontology, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
| | - Caroline S Fox
- The Framingham Heart Study, Framingham, MA, USA.,Division of Endocrinology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,The Framingham Heart Study, Framingham, MA, USA
| | - Philip A Wolf
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,The Framingham Heart Study, Framingham, MA, USA
| | - Ann C McKee
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,The Framingham Heart Study, Framingham, MA, USA
| | - Rhoda Au
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,The Framingham Heart Study, Framingham, MA, USA
| | - Lisa Delano-Wood
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Psychiatry, University of California, San Diego School of Medicine, La Jolla, CA, USA
| |
Collapse
|
104
|
Li W, Huang E. An Update on Type 2 Diabetes Mellitus as a Risk Factor for Dementia. J Alzheimers Dis 2018; 53:393-402. [PMID: 27163819 DOI: 10.3233/jad-160114] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
With the rapidly expanding evidence on brain structural and functional changes in type 2 diabetes mellitus (T2DM) patients, there is an increasing need to update our understanding on how T2DM associates with dementia as well as the underlying pathophysiological mechanisms. A literature search of T2DM and dementia or cognition impairments was carried out in electronic databases Medline, EMBASE, and Google Scholar. In this review, the chosen evidence was limited to human subject studies only, and data on either type 1 diabetes mellitus (T1DM) or non-classified diabetes were excluded. T2DM is a risk factor for both vascular dementia (VaD) and Alzheimer's disease (AD), although AD pathological marker studies have not provided sufficient evidence. T2DM interacts additively or synergistically with many factors, including old age, hypertension, total cholesterol, and APOEɛ4 carrier status for impaired cognition functions seen in patients with T2DM. In addition, comorbid T2DM can worsen the clinical presentations of patients with either AD or VaD. In summary, T2DM increases the risk for AD through different mechanisms for VaD although some mechanisms may overlap. Tau-related neurofibrillary tangles instead of amyloid-β plaques are more likely to be the pathological biomarkers for T2DM-related dementia. Degeneration of neurons in the brain, impaired regional blood supply/metabolism, and genetic predisposition are all involved in T2DM-associated dementia or cognitive impairments.
Collapse
Affiliation(s)
- Wei Li
- Master of Physician Assistant Studies, School of Health and Rehabilitation Sciences, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Edgar Huang
- School of Informatics and Computing, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| |
Collapse
|
105
|
Fatemi F, Kantarci K, Graff-Radford J, Preboske GM, Weigand SD, Przybelski SA, Knopman DS, Machulda MM, Roberts RO, Mielke MM, Petersen RC, Jack CR, Vemuri P. Sex differences in cerebrovascular pathologies on FLAIR in cognitively unimpaired elderly. Neurology 2018; 90:e466-e473. [PMID: 29343465 DOI: 10.1212/wnl.0000000000004913] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 10/27/2017] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE To examine sex differences in cerebrovascular pathologies (CVPs) as seen on fluid-attenuated inversion recovery (FLAIR) MRI and in cardiovascular and metabolic risk factors in a population-based cognitively unimpaired cohort and to examine whether sex is independently associated with FLAIR findings after accounting for differences in important midlife risk factors. METHODS We identified 1,301 cognitively normal participants (663 men and 638 women) enrolled in the Mayo Clinic Study of Aging (age ≥70 years) who had FLAIR MRI and ascertained total burden of white matter (WM) hyperintensities (WMH), subcortical infarctions, and cortical infarctions. We compared CVPs and midlife and late-life vascular risk factors between men and women. We fit regression models with each CVP as an outcome, treating age, sex, and midlife risk factors as predictors. RESULTS Women had significantly greater WMH volume relative to their WM volume compared to men (2.8% vs 2.4% of WM, p < 0.001), while men had a greater frequency of cortical infarctions compared to women (9% vs 4%, p < 0.001). Subcortical infarctions were equally common in men and women (20%). In regression modeling after adjustment for WM volume, the mean WMH volume difference between men and women was of the same magnitude as a 7-year difference in age. In contrast, men had 2.2-greater relative odds of having a cortical infarction compared to women. These sex differences persisted even after adjustment for midlife vascular risk factors. CONCLUSIONS There were important sex differences in CVP findings on FLAIR in cognitively unimpaired elderly. Understanding these sex differences could aid in the development of sex-specific preventive strategies.
Collapse
Affiliation(s)
- Farzan Fatemi
- From the Departments of Radiology (F.F., K.K., G.M.P., C.R.J., P.V.), Health Sciences Research (S.D.W., S.A.P., R.O.R., M.M. Mielke), Neurology (J.G.-R., D.S.K., R.O.R., M.M. Mielke, R.C.P.), and Psychology (M.M. Machulda), Mayo Clinic Rochester; and School of Medicine (F.F.), University of Minnesota, Minneapolis
| | - Kejal Kantarci
- From the Departments of Radiology (F.F., K.K., G.M.P., C.R.J., P.V.), Health Sciences Research (S.D.W., S.A.P., R.O.R., M.M. Mielke), Neurology (J.G.-R., D.S.K., R.O.R., M.M. Mielke, R.C.P.), and Psychology (M.M. Machulda), Mayo Clinic Rochester; and School of Medicine (F.F.), University of Minnesota, Minneapolis
| | - Jonathan Graff-Radford
- From the Departments of Radiology (F.F., K.K., G.M.P., C.R.J., P.V.), Health Sciences Research (S.D.W., S.A.P., R.O.R., M.M. Mielke), Neurology (J.G.-R., D.S.K., R.O.R., M.M. Mielke, R.C.P.), and Psychology (M.M. Machulda), Mayo Clinic Rochester; and School of Medicine (F.F.), University of Minnesota, Minneapolis
| | - Gregory M Preboske
- From the Departments of Radiology (F.F., K.K., G.M.P., C.R.J., P.V.), Health Sciences Research (S.D.W., S.A.P., R.O.R., M.M. Mielke), Neurology (J.G.-R., D.S.K., R.O.R., M.M. Mielke, R.C.P.), and Psychology (M.M. Machulda), Mayo Clinic Rochester; and School of Medicine (F.F.), University of Minnesota, Minneapolis
| | - Stephen D Weigand
- From the Departments of Radiology (F.F., K.K., G.M.P., C.R.J., P.V.), Health Sciences Research (S.D.W., S.A.P., R.O.R., M.M. Mielke), Neurology (J.G.-R., D.S.K., R.O.R., M.M. Mielke, R.C.P.), and Psychology (M.M. Machulda), Mayo Clinic Rochester; and School of Medicine (F.F.), University of Minnesota, Minneapolis
| | - Scott A Przybelski
- From the Departments of Radiology (F.F., K.K., G.M.P., C.R.J., P.V.), Health Sciences Research (S.D.W., S.A.P., R.O.R., M.M. Mielke), Neurology (J.G.-R., D.S.K., R.O.R., M.M. Mielke, R.C.P.), and Psychology (M.M. Machulda), Mayo Clinic Rochester; and School of Medicine (F.F.), University of Minnesota, Minneapolis
| | - David S Knopman
- From the Departments of Radiology (F.F., K.K., G.M.P., C.R.J., P.V.), Health Sciences Research (S.D.W., S.A.P., R.O.R., M.M. Mielke), Neurology (J.G.-R., D.S.K., R.O.R., M.M. Mielke, R.C.P.), and Psychology (M.M. Machulda), Mayo Clinic Rochester; and School of Medicine (F.F.), University of Minnesota, Minneapolis
| | - Mary M Machulda
- From the Departments of Radiology (F.F., K.K., G.M.P., C.R.J., P.V.), Health Sciences Research (S.D.W., S.A.P., R.O.R., M.M. Mielke), Neurology (J.G.-R., D.S.K., R.O.R., M.M. Mielke, R.C.P.), and Psychology (M.M. Machulda), Mayo Clinic Rochester; and School of Medicine (F.F.), University of Minnesota, Minneapolis
| | - Rosebud O Roberts
- From the Departments of Radiology (F.F., K.K., G.M.P., C.R.J., P.V.), Health Sciences Research (S.D.W., S.A.P., R.O.R., M.M. Mielke), Neurology (J.G.-R., D.S.K., R.O.R., M.M. Mielke, R.C.P.), and Psychology (M.M. Machulda), Mayo Clinic Rochester; and School of Medicine (F.F.), University of Minnesota, Minneapolis
| | - Michelle M Mielke
- From the Departments of Radiology (F.F., K.K., G.M.P., C.R.J., P.V.), Health Sciences Research (S.D.W., S.A.P., R.O.R., M.M. Mielke), Neurology (J.G.-R., D.S.K., R.O.R., M.M. Mielke, R.C.P.), and Psychology (M.M. Machulda), Mayo Clinic Rochester; and School of Medicine (F.F.), University of Minnesota, Minneapolis
| | - Ronald C Petersen
- From the Departments of Radiology (F.F., K.K., G.M.P., C.R.J., P.V.), Health Sciences Research (S.D.W., S.A.P., R.O.R., M.M. Mielke), Neurology (J.G.-R., D.S.K., R.O.R., M.M. Mielke, R.C.P.), and Psychology (M.M. Machulda), Mayo Clinic Rochester; and School of Medicine (F.F.), University of Minnesota, Minneapolis
| | - Clifford R Jack
- From the Departments of Radiology (F.F., K.K., G.M.P., C.R.J., P.V.), Health Sciences Research (S.D.W., S.A.P., R.O.R., M.M. Mielke), Neurology (J.G.-R., D.S.K., R.O.R., M.M. Mielke, R.C.P.), and Psychology (M.M. Machulda), Mayo Clinic Rochester; and School of Medicine (F.F.), University of Minnesota, Minneapolis
| | - Prashanthi Vemuri
- From the Departments of Radiology (F.F., K.K., G.M.P., C.R.J., P.V.), Health Sciences Research (S.D.W., S.A.P., R.O.R., M.M. Mielke), Neurology (J.G.-R., D.S.K., R.O.R., M.M. Mielke, R.C.P.), and Psychology (M.M. Machulda), Mayo Clinic Rochester; and School of Medicine (F.F.), University of Minnesota, Minneapolis.
| |
Collapse
|
106
|
Abstract
The American Diabetes Association (ADA) "Standards of Medical Care in Diabetes" includes ADA's current clinical practice recommendations and is intended to provide the components of diabetes care, general treatment goals and guidelines, and tools to evaluate quality of care. Members of the ADA Professional Practice Committee, a multidisciplinary expert committee, are responsible for updating the Standards of Care annually, or more frequently as warranted. For a detailed description of ADA standards, statements, and reports, as well as the evidence-grading system for ADA's clinical practice recommendations, please refer to the Standards of Care Introduction Readers who wish to comment on the Standards of Care are invited to do so at professional.diabetes.org/SOC.
Collapse
|
107
|
Kim J, Shin MS, Hwang SY, Park E, Lim YH, Shim JL, Kim SH, Kim YH, An M. Memory loss and decreased executive function are associated with limited functional capacity in patients with heart failure compared to patients with other medical conditions. Heart Lung 2018; 47:61-67. [DOI: 10.1016/j.hrtlng.2017.09.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 09/26/2017] [Accepted: 09/27/2017] [Indexed: 12/28/2022]
|
108
|
Ramanan VK, Przybelski SA, Graff-Radford J, Castillo AM, Lowe VJ, Mielke MM, Roberts RO, Reid RI, Knopman DS, Jack CR, Petersen RC, Vemuri P. Statins and Brain Health: Alzheimer's Disease and Cerebrovascular Disease Biomarkers in Older Adults. J Alzheimers Dis 2018; 65:1345-1352. [PMID: 30149450 PMCID: PMC6260813 DOI: 10.3233/jad-180446] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Statins have been proposed to reduce the risk of Alzheimer's disease (AD). OBJECTIVE Assess whether long-term statin use was associated with neuroimaging biomarkers of aging and dementia. METHODS Methods: We analyzed neuroimaging biomarkers in 1,160 individuals aged 65+ from the Mayo Clinic Study of Aging, a population-based prospective longitudinal study of cognitive aging. RESULTS Statin-treated (5+ years of therapy) individuals had greater burden of mid-and late-life cardiovascular disease (p < 0.001) than statin-untreated (≤3 months) individuals. Lower fractional anisotropy in the genu of the corpus callosum, an early marker of cerebrovascular disease, was associated with long-term statin exposure (p < 0.035). No significant associations were identified between long-term statin exposure and cerebral amyloid or tau burden, AD pattern neurodegeneration, or white matter hyperintensity burden. CONCLUSIONS Long-term statin therapy was not associated with differences in AD biomarkers. Individuals with long-term statin exposure had worse white matter integrity in the genu of the corpus callosum, consistent with the coexistence of higher cerebrovascular risk factor burden in this group.
Collapse
Affiliation(s)
- Vijay K Ramanan
- Department of Neurology, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| | - Scott A. Przybelski
- Department of Health Sciences Research, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| | | | - Anna M. Castillo
- Department of Health Sciences Research, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| | - Val J. Lowe
- Department of Radiology, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| | - Michelle M. Mielke
- Department of Neurology, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
- Department of Health Sciences Research, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| | - Rosebud O. Roberts
- Department of Neurology, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
- Department of Health Sciences Research, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| | - Robert I. Reid
- Department of Information Technology, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| | - David S. Knopman
- Department of Neurology, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| | - Clifford R. Jack
- Department of Radiology, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| | - Ronald C. Petersen
- Department of Neurology, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
- Department of Health Sciences Research, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| | - Prashanthi Vemuri
- Department of Radiology, Mayo Clinic-Rochester, Rochester, Minnesota, 55905, USA
| |
Collapse
|
109
|
Schneider ALC, Selvin E, Sharrett AR, Griswold M, Coresh J, Jack CR, Knopman D, Mosley T, Gottesman RF. Diabetes, Prediabetes, and Brain Volumes and Subclinical Cerebrovascular Disease on MRI: The Atherosclerosis Risk in Communities Neurocognitive Study (ARIC-NCS). Diabetes Care 2017; 40:1514-1521. [PMID: 28916531 PMCID: PMC5652590 DOI: 10.2337/dc17-1185] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/23/2017] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To examine the associations of prediabetes, diabetes, and diabetes severity (as assessed by HbA1c and diabetes duration) with brain volumes and vascular pathology on brain MRI and to assess whether the associations of diabetes with brain volumes are mediated by brain vascular pathology. RESEARCH DESIGN AND METHODS Cross-sectional study of 1,713 participants in the Atherosclerosis Risk in Communities Neurocognitive Study (ARIC-NCS) (mean age 75 years, 60% female, 27% black, 30% prediabetes, and 35% diabetes) who underwent 3T brain MRI scans in 2011-2013. Participants were categorized by diabetes-HbA1c status as without diabetes (<5.7% [reference]), with prediabetes (5.7 to <6.5%), and with diabetes ([defined as prior diagnosis or HbA1c ≥6.5%] <7.0% vs. ≥7.0%), with further stratification by diabetes duration (<10 vs. ≥10 years). RESULTS In adjusted analyses, compared with participants without diabetes and HbA1c <5.7%, participants with prediabetes and those with diabetes and HbA1c <7.0% did not have significantly different brain volumes or vascular pathology (all P > 0.05), but those with diabetes and HbA1c ≥7.0% had smaller total brain volume (β -0.20 SDs, 95% CI -0.31, -0.09), smaller regional brain volumes (including frontal, temporal, occipital, and parietal lobes; deep gray matter; Alzheimer disease signature region; and hippocampus [all P < 0.05]), and increased burden of white matter hyperintensities (WMH) (P = 0.016). Among participants with diabetes, those with HbA1c ≥7.0% had smaller total and regional brain volumes and an increased burden of WMH (all P < 0.05) compared with those with HbA1c <7.0%. Similarly, participants with longer duration of diabetes (≥10 years) had smaller brain volumes and higher burden of lacunes (all P < 0.05) than those with a diabetes duration <10 years. We found no evidence for mediation by WMH in associations of diabetes with smaller brain volumes by structural equation models (all P > 0.05). CONCLUSIONS More-severe diabetes (defined by higher HbA1c and longer disease duration) but not prediabetes or less-severe diabetes was associated with smaller brain volumes and an increased burden of brain vascular pathology. No evidence was found that associations of diabetes with smaller brain volumes are mediated by brain vascular pathology, suggesting that other mechanisms may be responsible for these associations.
Collapse
Affiliation(s)
- Andrea L C Schneider
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD .,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - A Richey Sharrett
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Michael Griswold
- Center of Biostatistics and Bioinformatics, University of Mississippi Medical Center, Jackson, MS
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | | | | | - Thomas Mosley
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS
| | - Rebecca F Gottesman
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
110
|
Byun MS, Kim HJ, Yi D, Choi HJ, Baek H, Lee JH, Choe YM, Sohn BK, Lee JY, Lee Y, Ko H, Kim YK, Lee YS, Sohn CH, Woo JI, Lee DY. Differential effects of blood insulin and HbA1c on cerebral amyloid burden and neurodegeneration in nondiabetic cognitively normal older adults. Neurobiol Aging 2017; 59:15-21. [DOI: 10.1016/j.neurobiolaging.2017.07.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 01/08/2023]
|
111
|
Macpherson H, Formica M, Harris E, Daly RM. Brain functional alterations in Type 2 Diabetes - A systematic review of fMRI studies. Front Neuroendocrinol 2017; 47:34-46. [PMID: 28687473 DOI: 10.1016/j.yfrne.2017.07.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 06/30/2017] [Accepted: 07/02/2017] [Indexed: 02/07/2023]
Abstract
Type 2 Diabetes (T2DM) is emerging as a major global health issue. T2DM can adversely affect cognition and increase dementia risk. This systematic review aimed to examine the functional brain changes that may underlie cognitive dysfunction in adults with T2DM. Studies were restricted to those which used functional magnetic resonance imaging (fMRI). Nineteen independent studies were identified, mostly comprised of middle aged or older adults. Resting-state studies demonstrated that compared to controls, connectivity of the Default Mode Network (DMN) was reduced and the majority of task-based studies identified reduced activation in T2DM patients in regions relevant to task performance. Abnormalities of low frequency spontaneous brain activity were observed, particularly in visual regions. As most studies demonstrated that alterations in fMRI were related to poorer neuropsychological task performance, these results indicate that functional brain abnormalities in T2DM have consequences for cognition.
Collapse
Affiliation(s)
- Helen Macpherson
- Institute for Physical Activity and Nutrition, Deakin University, Geelong, VIC, Australia.
| | - Melissa Formica
- Institute for Physical Activity and Nutrition, Deakin University, Geelong, VIC, Australia
| | - Elizabeth Harris
- Centre for Human Psychopharmacology, Swinburne University, Hawthorn, VIC, Australia
| | - Robin M Daly
- Institute for Physical Activity and Nutrition, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
112
|
Bidirectional interactions between diabetes and Alzheimer's disease. Neurochem Int 2017; 108:296-302. [PMID: 28551028 DOI: 10.1016/j.neuint.2017.04.020] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 04/24/2017] [Accepted: 04/27/2017] [Indexed: 01/21/2023]
|
113
|
Hyperglycemia induced the Alzheimer's proteins and promoted loss of synaptic proteins in advanced-age female Goto-Kakizaki (GK) rats. Neurosci Lett 2017; 655:41-45. [PMID: 28652187 DOI: 10.1016/j.neulet.2017.06.041] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Revised: 06/08/2017] [Accepted: 06/23/2017] [Indexed: 11/21/2022]
Abstract
Although both type 2 diabetes mellitus (T2DM) and aging are related with Alzheimer's disease (AD), the effects of aging on the Alzheimer's proteins and the synaptic markers in T2DM have not been investigated. This study, we hypothesized that T2DM rats with advanced-age, aggravates the reduction of synaptic proteins and an increase in the Alzheimer's protein markers. Goto-Kakizaki rats (GK) were used as a T2DM group and wild-type rats (WT) were used as a control group. Rats in each group were categorized by age into young-adult (7 months) and advanced-age rats (12.5 months). Blood was collected in all rats to determine plasma glucose and insulin levels. The brains were used for determining the level of Alzheimer's and synaptic proteins. Our data demonstrated that GK rats had a decreased body weight and increased blood glucose levels, compared to their age-matched WT. p-Tau was increased in both advanced-age WT and GK, compared to their young-adult rats. Moreover, amyloid-beta (Aβ) level was higher in advanced-age GK than their age-matched WT. The synaptic proteins were decreased in advanced-age GK, compared to young-adult GK rats. However, no difference in the level of Alzheimer's proteins and synaptic proteins in the brains of young-adult GK compared to age-matched WT was found. Our data suggested that aging contributes to the pathogenesis of AD and the reduction of synaptic proteins to greater extent in a diabetic than in a healthy condition.
Collapse
|
114
|
Ong M, Foo H, Chander RJ, Wen MC, Au WL, Sitoh YY, Tan L, Kandiah N. Influence of diabetes mellitus on longitudinal atrophy and cognition in Parkinson's disease. J Neurol Sci 2017; 377:122-126. [DOI: 10.1016/j.jns.2017.04.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/22/2017] [Accepted: 04/10/2017] [Indexed: 11/30/2022]
|
115
|
Vemuri P, Knopman DS, Lesnick TG, Przybelski SA, Mielke MM, Graff-Radford J, Murray ME, Roberts RO, Vassilaki M, Lowe VJ, Machulda MM, Jones DT, Petersen RC, Jack CR. Evaluation of Amyloid Protective Factors and Alzheimer Disease Neurodegeneration Protective Factors in Elderly Individuals. JAMA Neurol 2017; 74:718-726. [PMID: 28418521 PMCID: PMC5649401 DOI: 10.1001/jamaneurol.2017.0244] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 02/28/2017] [Indexed: 01/11/2023]
Abstract
Importance While amyloid and neurodegeneration are viewed together as Alzheimer disease pathophysiology (ADP), the factors that influence amyloid and AD-pattern neurodegeneration may be considerably different. Protection from these ADP factors may be important for aging without significant ADP. Objective To identify the combined and independent protective factors for amyloid and AD-pattern neurodegeneration in a population-based sample and to test the hypothesis that "exceptional agers" with advanced ages do not have significant ADP because they have protective factors for amyloid and neurodegeneration. Design, Setting, and Participants This cohort study conducted a prospective analysis of 942 elderly individuals (70-≥90 years) with magnetic resonance imaging and Pittsburgh compound B-positron emission tomography scans enrolled in the Mayo Clinic Study of Aging, a longitudinal population-based study of cognitive aging in Olmsted County, Minnesota. We operationalized "exceptional aging" without ADP by considering individuals 85 years or older to be without significant evidence of ADP. Main Outcomes and Measures We evaluated predictors including demographics, APOE, intellectual enrichment, midlife risk factors (physical inactivity, obesity, smoking, diabetes, hypertension, and dyslipidemia), and the total number of late-life cardiac and metabolic conditions. We used multivariate linear regression models to identify the combined and independent protective factors for amyloid and AD-pattern neurodegeneration. Using a subsample of the cohort 85 years of age or older, we computed Cohen d-based effect size estimations to compare the quantitative strength of each predictor variable in their contribution with exceptional aging without ADP. Results The study participants included 423 (45%) women and the average age of participants was 79.7 (5.9) years. Apart from demographics and the APOE genotype, only midlife dyslipidemia was associated with amyloid deposition. Obesity, smoking, diabetes, hypertension, and cardiac and metabolic conditions, but not intellectual enrichment, were associated with greater AD-pattern neurodegeneration. In the 85 years or older cohort, the Cohen d results showed small to moderate effects (effect sizes > 0.2) of several variables except job score and midlife hypertension in predicting exceptional aging without ADP. Conclusions and Relevance The protective factors that influence amyloid and AD-pattern neurodegeneration are different. "Exceptional aging" without ADP may be possible with a greater number of protective factors across the lifespan but warrants further investigation.
Collapse
Affiliation(s)
- Prashanthi Vemuri
- Department of Radiology, Mayo Clinic–Rochester, Rochester, Minnesota
| | - David S. Knopman
- Department of Neurology, Mayo Clinic–Rochester, Rochester, Minnesota
| | - Timothy G. Lesnick
- Department of Health Sciences Research, Mayo Clinic–Rochester, Rochester, Minnesota
| | - Scott A. Przybelski
- Department of Health Sciences Research, Mayo Clinic–Rochester, Rochester, Minnesota
| | - Michelle M. Mielke
- Department of Health Sciences Research, Mayo Clinic–Rochester, Rochester, Minnesota
| | | | - Melissa E. Murray
- Department of Neuroscience, Mayo Clinic–Jacksonville, Jacksonville, Florida
| | - Rosebud O. Roberts
- Department of Health Sciences Research, Mayo Clinic–Rochester, Rochester, Minnesota
| | - Maria Vassilaki
- Department of Health Sciences Research, Mayo Clinic–Rochester, Rochester, Minnesota
| | - Val J. Lowe
- Department of Radiology, Mayo Clinic–Rochester, Rochester, Minnesota
| | - Mary M. Machulda
- Department of Psychology, Mayo Clinic–Rochester, Rochester, Minnesota
| | - David T. Jones
- Department of Neurology, Mayo Clinic–Rochester, Rochester, Minnesota
| | | | - Clifford R. Jack
- Department of Radiology, Mayo Clinic–Rochester, Rochester, Minnesota
| |
Collapse
|
116
|
Eberhardt O, Topka H. Neurological outcomes of antidiabetic therapy: What the neurologist should know. Clin Neurol Neurosurg 2017; 158:60-66. [PMID: 28477558 DOI: 10.1016/j.clineuro.2017.04.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 03/05/2017] [Accepted: 04/15/2017] [Indexed: 02/09/2023]
Abstract
Considering the causative or contributory effects of diabetes mellitus on common neurological diseases such as polyneuropathy, stroke and dementia, modern antidiabetic drugs may be expected to reduce incidence or progression of these conditions. Nevertheless, most observed benefits have been small, except in the context of therapy for diabetes mellitus type I and new-onset polyneuropathy. Recently, semaglutide, a GLP-1 analog, has been shown to significantly reduce stroke incidence in a randomized controlled trial. Beneficial effects of antidiabetic drugs on stroke severity or outcome have been controversial, though. The level of risk conferred by diabetes mellitus, the complex pathophysiology of neurological diseases, issues of trial design, side-effects of antidiabetic drugs as well as co-medication might be interacting factors that determine the performance of antidiabetic therapy with respect to neurological outcomes. It might be speculated that early treatment of prediabetes might prevent cerebral arteriosclerosis, cognitive decline or polyneuropathy more effectively, but this remains to be demonstrated.
Collapse
Affiliation(s)
- Olaf Eberhardt
- Department for Neurology, Clinical Neurophysiology, Clinical Neuropsychology and Stroke Unit, Klinikum Bogenhausen Englschalkinger Str. 77, München, 81925, Germany.
| | - Helge Topka
- Department for Neurology, Clinical Neurophysiology, Clinical Neuropsychology and Stroke Unit, Klinikum Bogenhausen Englschalkinger Str. 77, München, 81925, Germany
| |
Collapse
|
117
|
Contribution of thrombin-reactive brain pericytes to blood-brain barrier dysfunction in an in vivo mouse model of obesity-associated diabetes and an in vitro rat model. PLoS One 2017; 12:e0177447. [PMID: 28489922 PMCID: PMC5425209 DOI: 10.1371/journal.pone.0177447] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 04/27/2017] [Indexed: 12/14/2022] Open
Abstract
Diabetic complications are characterized by the dysfunction of pericytes located around microvascular endothelial cells. The blood–brain barrier (BBB) exhibits hyperpermeability with progression of diabetes. Therefore, brain pericytes at the BBB may be involved in diabetic complications of the central nervous system (CNS). We hypothesized that brain pericytes respond to increased brain thrombin levels in diabetes, leading to BBB dysfunction and diabetic CNS complications. Mice were fed a high-fat diet (HFD) for 2 or 8 weeks to induce obesity. Transport of i.v.-administered sodium fluorescein and 125I-thrombin across the BBB were measured. We evaluated brain endothelial permeability and expression of tight junction proteins in the presence of thrombin–treated brain pericytes using a BBB model of co-cultured rat brain endothelial cells and pericytes. Mice fed a HFD for 8 weeks showed both increased weight gain and impaired glucose tolerance. In parallel, the brain influx rate of sodium fluorescein was significantly greater than that in mice fed a normal diet. HFD feeding inhibited the decline in brain thrombin levels occurring during 6 weeks of feeding. In the HFD fed mice, plasma thrombin levels were significantly increased, by up to 22%. 125I-thrombin was transported across the BBB in normal mice after i.v. injection, with uptake further enhanced by co-injection of unlabeled thrombin. Thrombin-treated brain pericytes increased brain endothelial permeability and caused decreased expression of zona occludens-1 (ZO-1) and occludin and morphological disorganization of ZO-1. Thrombin also increased mRNA expression of interleukin-1β and 6 and tumor necrosis factor-α in brain pericytes. Thrombin can be transported from circulating blood through the BBB, maintaining constant levels in the brain, where it can stimulate pericytes to induce BBB dysfunction. Thus, the brain pericyte–thrombin interaction may play a key role in causing BBB dysfunction in obesity-associated diabetes and represent a therapeutic target for its CNS complications.
Collapse
|
118
|
Haghir H, Hami J, Lotfi N, Peyvandi M, Ghasemi S, Hosseini M. Expression of apoptosis-regulatory genes in the hippocampus of rat neonates born to mothers with diabetes. Metab Brain Dis 2017; 32:617-628. [PMID: 28078553 DOI: 10.1007/s11011-017-9950-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 01/03/2017] [Indexed: 01/01/2023]
Abstract
Diabetes during pregnancy impairs the development of the central nervous system (CNS) and causes cognitive and behavioral abnormalities in offspring. However, the exact mechanism by which the maternal diabetes affects the development of the brain remains to be elucidated. The aim of the present study was to investigate the effects of maternal diabetes in pregnancy on the expression of Bcl-2 and Bax genes and the numerical density of degenerating dark neurons (DNs) in the hippocampus of offspring at the first postnatal two weeks. Wistar female rats were maintained diabetic from a week before pregnancy through parturition and male offspring was sacrificed at P0, P7, and P14. Our findings demonstrated a significant down-regulation in the hippocampal expression of Bcl-2 in the diabetic group newborns (P < 0.05). In contrast, the mRNA expression of Bax was markedly up-regulated in the offspring born to diabetic dams at all of studied time-points (P < 0.05). Moreover, we found a striking increase in the numerical density of DNs in the various subfields of hippocampus of diabetic group pups (P < 0.05). The results of the present study revealed that maternal hyperglycemia during gestational period may result in disturbances in the expression of Bcl-2 and Bax genes as two important genes in neuronal apoptosis regulation and induces the production of DNs in the developing hippocampus of neonatal rats. These disturbances may be a reason for the cognitive, structural, and behavioral anomalies observed in offspring born to diabetic mothers. Furthermore, the control of maternal glycaemia by insulin administration in most cases normalized these negative impacts.
Collapse
Affiliation(s)
- Hossein Haghir
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetic Research Center (MGRC), School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Hami
- Department of Anatomical Sciences, School of Medicine, Birjand University of Medical Sciences, Ghaffari St., Birjand, Iran.
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| | - Nassim Lotfi
- Department of Anatomical Sciences, School of Medicine, Birjand University of Medical Sciences, Ghaffari St., Birjand, Iran
| | - Mostafa Peyvandi
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Simagol Ghasemi
- Microanatomy Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehran Hosseini
- Department of Public Health, Deputy of Research and Technology, Research Centre of Experimental Medicine, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
119
|
Reitz C, Guzman VA, Narkhede A, DeCarli C, Brickman AM, Luchsinger JA. Relation of Dysglycemia to Structural Brain Changes in a Multiethnic Elderly Cohort. J Am Geriatr Soc 2017; 65:277-285. [PMID: 27917464 PMCID: PMC5311018 DOI: 10.1111/jgs.14551] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Abnormally high glucose levels (dysglycemia) increase with age. Epidemiological studies suggest that dysglycemia is a risk factor for cognitive impairment but the underlying pathophysiological mechanisms remain unclear. The objective of this study was to examine the relation of dysglycemia clinical categories (normal glucose tolerance (NGT), pre-diabetes, undiagnosed diabetes, known diabetes) with brain structure in older adults. We also assessed the relation between dysglycemia and cognitive performance. DESIGN Cross-sectional and longitudinal cohort study. SETTING Northern Manhattan (Washington Heights, Hamilton Heights, and Inwood). PARTICIPANTS Medicare recipients 65 years and older. MEASUREMENTS Dysglycemia categories were based on HBA1c or history of type 2 diabetes (diabetes). Brain structure (brain infarcts, white matter hyperintensities (WMH) volume, total gray matter volume, total white matter volume, total hippocampus volume) was assessed with brain magnetic resonance imaging; cognitive function (memory, language and visuospatial function, speed) was assessed with a validated neuropsychological battery. RESULTS Dysglycemia, defined with HbA1c as a continuous variable or categorically as pre-diabetes and diabetes, was associated with a higher number of brain infarcts, WMH volume and decreased total white matter, gray matter and hippocampus volumes cross-sectionally, and a significant decline in gray matter volume longitudinally. Dysglycemia was also associated with lower performance in language, speed and visuospatial function although these associations were attenuated when adjusted for education, APOE-ε4, ethnic group and vascular risk factors. CONCLUSION Our results suggest that dysglycemia affects brain structure and cognition even in elderly survivors, evidenced by higher cerebrovascular disease, lower white and gray matter volume, and worse language and visuospatial function and cognitive speed.
Collapse
Affiliation(s)
- Christiane Reitz
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY
| | - Vanessa A. Guzman
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Atul Narkhede
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Charles DeCarli
- Department of Neurology, Center for Neuroscience, University of California at Davis, Sacramento
- Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis, Sacramento
| | - Adam M. Brickman
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, College of Physicians and Surgeons, Columbia University, New York, NY
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY
| | - José A. Luchsinger
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY
| |
Collapse
|
120
|
Time-Dependent Lactate Production and Amino Acid Utilization in Cultured Astrocytes Under High Glucose Exposure. Mol Neurobiol 2017; 55:1112-1122. [DOI: 10.1007/s12035-016-0360-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 12/28/2016] [Indexed: 02/07/2023]
|
121
|
|
122
|
Zheng H, Zheng Y, Wang D, Cai A, Lin Q, Zhao L, Chen M, Deng M, Ye X, Gao H. Analysis of neuron-astrocyte metabolic cooperation in the brain of db/db mice with cognitive decline using 13C NMR spectroscopy. J Cereb Blood Flow Metab 2017; 37:332-343. [PMID: 26762505 PMCID: PMC5363750 DOI: 10.1177/0271678x15626154] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 12/01/2015] [Accepted: 12/07/2015] [Indexed: 01/24/2023]
Abstract
Type 2 diabetes has been linked to cognitive impairment, but its potential metabolic mechanism is still unclear. The present study aimed to explore neuron-astrocyte metabolic cooperation in the brain of diabetic (db/db, BKS.Cg-m+/+ Leprdb/J) mice with cognitive decline using 13C NMR technique in combination with intravenous [2-13C]-acetate and [3-13C]-lactate infusions. We found that the 13C-enrichment from [2-13C]-acetate into tricarboxylic acid cycle intermediate, succinate, was significantly decreased in db/db mice with cognitive decline compared with wild-type (WT, C57BLKS/J) mice, while an opposite result was obtained after [3-13C]-lactate infusion. Relative to WT mice, db/db mice with cognitive decline had significantly lower 13C labeling percentages in neurotransmitters including glutamine, glutamate, and γ-aminobutyric acid after [2-13C]-acetate infusion. However, [3-13C]-lactate resulted in increased 13C-enrichments in neurotransmitters in db/db mice with cognitive decline. This may indicate that the disturbance of neurotransmitter metabolism occurred during the development of cognitive decline. In addition, a reduction in 13C-labeling of lactate and an increase in gluconeogenesis were found from both labeled infusions in db/db mice with cognitive decline. Therefore, our results suggest that the development of cognitive decline in type 2 diabetes may be implicated to an unbalanced metabolism in neuron-astrocyte cooperation and an enhancement of gluconeogenesis.
Collapse
Affiliation(s)
- Hong Zheng
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yongquan Zheng
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,Radiology Department of the Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Dan Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Aimin Cai
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Qiuting Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Liangcai Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Minjiang Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Mingjie Deng
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xinjian Ye
- Radiology Department of the Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Hongchang Gao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
123
|
Raman MR, Kantarci K, Murray ME, Jack CR, Vemuri P. Imaging markers of cerebrovascular pathologies: Pathophysiology, clinical presentation, and risk factors. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2016; 5:5-14. [PMID: 28054023 PMCID: PMC5198884 DOI: 10.1016/j.dadm.2016.12.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cerebrovascular pathologies (CVPs) are common pathologies associated with age-related cognitive decline along with Alzheimer disease pathologies. The impact of CVP on the prevalence of dementia is increasingly being recognized. The goal of this review is to improve our understanding of the pathophysiological underpinnings and the multimodal magnetic resonance imaging and positron emission tomography imaging changes that are associated with the hallmarks of CVP. This knowledge will facilitate the development of early detection, intervention, and prevention strategies that may contribute to lowering the risk of dementia. In this review, we will first discuss currently known risk factors of CVPs including cardiovascular, lifestyle, genetic, sex differences, and head injury. Next, we will focus on the pathophysiology of CVPs and their impact on neurodegeneration and downstream cognitive impairment. Specifically, we will discuss three of the most common cerebrovascular lesions seen on MRI: white-matter hyperintensity, microbleeds, and infarcts. Finally, we will discuss the unanswered open questions in this field.
Collapse
Affiliation(s)
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | | | | |
Collapse
|
124
|
Safarpour D, Willis AW. Clinical Epidemiology, Evaluation, and Management of Dementia in Parkinson Disease. Am J Alzheimers Dis Other Demen 2016; 31:585-594. [PMID: 27295974 PMCID: PMC10852884 DOI: 10.1177/1533317516653823] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The prevalence of neurodegenerative diseases such as Parkinson disease (PD) will increase substantially, due to the aging of the population and improved treatments leading to better disease-related outcomes. Dementia is the most common nonmotor symptom in PD, and most patients with PD will have cognitive dysfunction and cognitive decline in the course of their disease. The development of cognitive dysfunction in PD greatly limits the ability to participate in activities of daily living and can be a tipping point for nursing home placement or major caregiver stress. Understanding the different causes of dementia and how to reduce the incidence and impact of secondary cognitive dysfunction in PD are necessary skills for primary care physicians and neurologists. In this review, we discuss the clinical epidemiology of dementia in PD with an emphasis on preventable cognitive dysfunction, present tools for outpatient evaluation of cognitive dysfunction, and describe current pharmacological treatments for dementia in PD.
Collapse
Affiliation(s)
- Delaram Safarpour
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA Parkinson's Disease Research, Education and Clinical Center, Philadelphia VA Medical Center, Philadelphia, PA, USA
| | - Allison W Willis
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA Department of Biostatistics and Epidemiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA Leonard Davis Institute of Health Economics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
125
|
Hendrie HC, Zheng M, Li W, Lane K, Ambuehl R, Purnell C, Unverzagt FW, Torke A, Balasubramanyam A, Callahan CM, Gao S. Glucose level decline precedes dementia in elderly African Americans with diabetes. Alzheimers Dement 2016; 13:111-118. [PMID: 27793691 DOI: 10.1016/j.jalz.2016.08.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 08/26/2016] [Indexed: 12/31/2022]
Abstract
INTRODUCTION High blood glucose levels may be responsible for the increased risk for dementia in diabetic patients. METHODS A secondary data analysis merging electronic medical records (EMRs) with data collected from the Indianapolis-Ibadan Dementia project (IIDP). Of the enrolled 4105 African Americans, 3778 were identified in the EMR. Study endpoints were dementia, mild cognitive impairment (MCI), or normal cognition. Repeated serum glucose measurements were used as the outcome variables. RESULTS Diabetic participants who developed incident dementia had a significant decrease in serum glucose levels in the years preceding the diagnosis compared to the participants with normal cognition (P = .0002). They also had significantly higher glucose levels up to 9 years before the dementia diagnosis (P = .0367). DISCUSSION High glucose levels followed by a decline occurring years before diagnosis in African American participants with diabetes may represent a powerful presymptomatic metabolic indicator of dementia.
Collapse
Affiliation(s)
- Hugh C Hendrie
- Indiana University Center for Aging Research, Indianapolis, IN, USA; Regenstrief Institute, Inc., Indianapolis, IN, USA; Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Mengjie Zheng
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Wei Li
- School of Health and Rehabilitation Sciences, IUPUI, Indianapolis, IN, USA
| | - Kathleen Lane
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | - Frederick W Unverzagt
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alexia Torke
- Indiana University Center for Aging Research, Indianapolis, IN, USA; Regenstrief Institute, Inc., Indianapolis, IN, USA; Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Chris M Callahan
- Indiana University Center for Aging Research, Indianapolis, IN, USA; Regenstrief Institute, Inc., Indianapolis, IN, USA; Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sujuan Gao
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
126
|
McArthur S, Loiola RA, Maggioli E, Errede M, Virgintino D, Solito E. The restorative role of annexin A1 at the blood-brain barrier. Fluids Barriers CNS 2016; 13:17. [PMID: 27655189 PMCID: PMC5031267 DOI: 10.1186/s12987-016-0043-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 09/12/2016] [Indexed: 12/20/2022] Open
Abstract
Annexin A1 is a potent anti-inflammatory molecule that has been extensively studied in the peripheral immune system, but has not as yet been exploited as a therapeutic target/agent. In the last decade, we have undertaken the study of this molecule in the central nervous system (CNS), focusing particularly on the primary interface between the peripheral body and CNS: the blood-brain barrier. In this review, we provide an overview of the role of this molecule in the brain, with a particular emphasis on its functions in the endothelium of the blood-brain barrier, and the protective actions the molecule may exert in neuroinflammatory, neurovascular and metabolic disease. We focus on the possible new therapeutic avenues opened up by an increased understanding of the role of annexin A1 in the CNS vasculature, and its potential for repairing blood-brain barrier damage in disease and aging.
Collapse
Affiliation(s)
- Simon McArthur
- Department of Biomedical Sciences, Faculty of Science and Technology, University of Westminster, London, UK
| | - Rodrigo Azevedo Loiola
- William Harvey Research Institute, School of Medicine and Dentistry, Queen Mary University, London, UK
| | - Elisa Maggioli
- William Harvey Research Institute, School of Medicine and Dentistry, Queen Mary University, London, UK
| | - Mariella Errede
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Bari University School of Medicine, Bari, Italy
| | - Daniela Virgintino
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, Bari University School of Medicine, Bari, Italy
| | - Egle Solito
- William Harvey Research Institute, School of Medicine and Dentistry, Queen Mary University, London, UK
| |
Collapse
|
127
|
Hirabayashi N, Hata J, Ohara T, Mukai N, Nagata M, Shibata M, Gotoh S, Furuta Y, Yamashita F, Yoshihara K, Kitazono T, Sudo N, Kiyohara Y, Ninomiya T. Association Between Diabetes and Hippocampal Atrophy in Elderly Japanese: The Hisayama Study. Diabetes Care 2016; 39:1543-9. [PMID: 27385328 DOI: 10.2337/dc15-2800] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/16/2016] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To investigate the association between diabetes and brain or hippocampal atrophy in an elderly population. RESEARCH DESIGN AND METHODS A total of 1,238 community-dwelling Japanese subjects aged ≥65 years underwent brain MRI scans and a comprehensive health examination in 2012. Total brain volume (TBV), intracranial volume (ICV), and hippocampal volume (HV) were measured using MRI scans for each subject. We examined the associations between diabetes-related parameters and the ratios of TBV to ICV (an indicator of global brain atrophy), HV to ICV (an indicator of hippocampal atrophy), and HV to TBV (an indicator of hippocampal atrophy beyond global brain atrophy) after adjustment for other potential confounders. RESULTS The multivariable-adjusted mean values of the TBV-to-ICV, HV-to-ICV, and HV-to-TBV ratios were significantly lower in the subjects with diabetes compared with those without diabetes (77.6% vs. 78.2% for the TBV-to-ICV ratio, 0.513% vs. 0.529% for the HV-to-ICV ratio, and 0.660% vs. 0.676% for the HV-to-TBV ratio; all P < 0.01). These three ratios decreased significantly with elevated 2-h postload glucose (PG) levels (all P for trend <0.05) but not fasting plasma glucose levels. Longer duration of diabetes was significantly associated with lower TBV-to-ICV, HV-to-ICV, and HV-to-TBV ratios. The subjects with diabetes diagnosed in midlife had significantly lower HV-to-ICV and HV-to-TBV ratios than those without and those diagnosed in late life. CONCLUSIONS Our data suggest that a longer duration of diabetes and elevated 2-h PG levels, a marker of postprandial hyperglycemia, are risk factors for brain atrophy, particularly hippocampal atrophy.
Collapse
Affiliation(s)
- Naoki Hirabayashi
- Department of Environmental Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan Department of Psychosomatic Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jun Hata
- Department of Environmental Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoyuki Ohara
- Department of Environmental Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Naoko Mukai
- Department of Environmental Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaharu Nagata
- Department of Environmental Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mao Shibata
- Department of Environmental Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan Department of Psychosomatic Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Seiji Gotoh
- Department of Environmental Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiko Furuta
- Department of Environmental Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Fumio Yamashita
- Division of Ultrahigh Field MRI, Institute for Biomedical Sciences, Iwate Medical University, Iwate, Japan
| | - Kazufumi Yoshihara
- Department of Psychosomatic Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takanari Kitazono
- Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Nobuyuki Sudo
- Department of Psychosomatic Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yutaka Kiyohara
- Department of Environmental Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshiharu Ninomiya
- Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
128
|
Umemura T, Kawamura T, Hotta N. Pathogenesis and neuroimaging of cerebral large and small vessel disease in type 2 diabetes: A possible link between cerebral and retinal microvascular abnormalities. J Diabetes Investig 2016; 8:134-148. [PMID: 27239779 PMCID: PMC5334292 DOI: 10.1111/jdi.12545] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 05/23/2016] [Indexed: 01/14/2023] Open
Abstract
Diabetes patients have more than double the risk of ischemic stroke compared with non‐diabetic individuals, and its neuroimaging characteristics have important clinical implications. To understand the pathophysiology of ischemic stroke in diabetes, it is important to focus not only on the stroke subtype, but also on the size and location of the occlusive vessels. Specifically, ischemic stroke in diabetes patients might be attributed to both large and small vessels, and intracranial internal carotid artery disease and small infarcts of the posterior circulation often occur. An additional feature is that asymptomatic lacunar infarctions are often seen in the basal ganglia and brain stem on brain magnetic resonance imaging. In particular, cerebral small vessel disease (SVD), including lacunar infarctions, white matter lesions and cerebral microbleeds, has been shown to be associated not only with stroke incidence, but also with the development and progression of dementia and diabetic microangiopathy. However, the pathogenesis of cerebral SVD is not fully understood. In addition, data on the association between neuroimaging findings of the cerebral SVD and diabetes are limited. Recently, the clinical importance of the link between cerebral SVD and retinal microvascular abnormalities has been a topic of considerable interest. Several clinical studies have shown that retinal microvascular abnormalities are closely related to cerebral SVD, suggesting that retinal microvascular abnormalities might be pathophysiologically linked to ischemic cerebral SVD. We review the literature relating to the pathophysiology and neuroimaging of cerebrovascular disease in diabetes, and discuss the problems based on the concept of cerebral large and small vessel disease.
Collapse
Affiliation(s)
| | - Takahiko Kawamura
- Department of Diabetes and Endocrine Internal Medicine, Chubu Rosai Hospital, Nagoya, Japan.,Department of Preventive Medical Center, Chubu Rosai Hospital, Nagoya, Japan
| | - Nigishi Hotta
- Department of Diabetes and Endocrine Internal Medicine, Chubu Rosai Hospital, Nagoya, Japan
| |
Collapse
|
129
|
Huang H, Nie S, Cao M, Marshall C, Gao J, Xiao N, Hu G, Xiao M. Characterization of AD-like phenotype in aged APPSwe/PS1dE9 mice. AGE (DORDRECHT, NETHERLANDS) 2016; 38:303-322. [PMID: 27439903 PMCID: PMC5061676 DOI: 10.1007/s11357-016-9929-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 07/12/2016] [Indexed: 05/28/2023]
Abstract
Transgenic APPSwe/PS1dE9 (APP/PS1) mice that overproduce amyloid beta (Aβ) are extensively used in the studies of pathogenesis and experimental therapeutics and new drug screening for Alzheimer's disease (AD). However, most of the current literature uses young or adult APP/PS1 mice. In order to provide a broader view of AD-like phenotype of this animal model, in this study, we systematically analyzed behavioral and pathological profiles of 24-month-old male APP/PS1 mice. Aged APP/PS1 mice had reference memory deficits as well as anxiety, hyperactivity, and social interaction impairment. Consistently, there was obvious deposition of amyloid plaques in the dorsal hippocampus with decreased expression of insulin-degrading enzyme, a proteolytic enzyme responsible for degradation of intracellular Aβ. Furthermore, decreases in hippocampal volume, neuronal number and synaptophysin expression, and astrocyte atrophy were also observed in aged APP/PS1 mice. This finding suggests that aged APP/PS1 mice can well replicate cognitive and noncognitive behavioral abnormalities, hippocampal atrophy, and neuronal and astrocyte degeneration in AD patients, to enable more objective and refined preclinical evaluation of therapeutic drugs and strategies for AD treatment.
Collapse
Affiliation(s)
- Huang Huang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, Jiangsu, 211166, China
- Department of Neurology, Sir Run Run Shaw Hospital, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Sipei Nie
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, Jiangsu, 211166, China
| | - Min Cao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, Jiangsu, 211166, China
| | - Charles Marshall
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, Jiangsu, 211166, China
- Department of Rehabilitation Sciences, University of Kentucky Center of Excellence in Rural Health, Hazard, KY, 41701, USA
| | - Junying Gao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, Jiangsu, 211166, China
| | - Na Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, Jiangsu, 211166, China
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, Jiangsu, 211166, China
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
130
|
Hilal S, Amin SM, Venketasubramanian N, Niessen WJ, Vrooman H, Wong TY, Chen C, Ikram MK. Subcortical Atrophy in Cognitive Impairment and Dementia. J Alzheimers Dis 2016; 48:813-23. [PMID: 26402115 DOI: 10.3233/jad-150473] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Cortical atrophy is a key neuroimaging feature of dementia. However, the role of subcortical gray matter reduction in cognitive impairment has not been explored extensively. OBJECTIVES We examined the risk factors of subcortical structures on neuroimaging and their association with cognitive impairment and dementia. METHODS Data from two studies were used: a subsample from the Epidemiology of Dementia in Singapore (EDIS) study of non-demented community-dwelling subjects (n = 550) and a case-control study. Subjects underwent similar neuropsychological tests and brain MRI. Subcortical volumes of accumbens, amygdala, caudate, pallidum, putamen, thalamus, hippocampus, and brainstem were measured. Cognitive impairment no dementia (CIND), dementia and its subtypes, vascular cognitive impairment (VCI), were defined using accepted criteria. Cognitive function was also expressed as both composite and domain-specific Z-scores. RESULTS In the EDIS study, age, female gender, Malay ethnicity, diabetes, lacunar-infarcts, and white matter lesions were the most important risk factors for subcortical atrophy. Moreover, smaller volumes of accumbens, amygdala, caudate, thalamus, and brainstem were significantly associated with lower cognitive composite Z-scores. With respect to clinical outcomes in the case-control study, structures such as the accumbens, caudate, putamen, and hippocampus were associated with both CIND and dementia. Smaller caudate and pallidum volumes were related to VCI whereas amygdalar atrophy was only associated with non-VCI. Furthermore, subcortical atrophy was related to both VCI and non-VCI. CONCLUSION Subcortical gray matter atrophy is not only observed in dementia, but also in the preclinical stages of cognitive impairment. Furthermore, besides VCI, subcortical structures were also related to non-VCI.
Collapse
Affiliation(s)
- Saima Hilal
- Memory Ageing and Cognition Centre (MACC), National University Health System, Singapore.,Department of Pharmacology, National University of Singapore, Singapore
| | - Shaik Muhammad Amin
- Memory Ageing and Cognition Centre (MACC), National University Health System, Singapore.,Department of Pharmacology, National University of Singapore, Singapore
| | | | - Wiro J Niessen
- Departments of Radiology & Medical Informatics, Erasmus University Medical Center, Rotterdam, The Netherlands.,Faculty of Applied Sciences, Delft University of Technology, The Netherlands
| | - Henri Vrooman
- Departments of Radiology & Medical Informatics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Center, Singapore.,Academic Medicine Research Institute, Duke-NUS Graduate Medical School, Singapore
| | - Christopher Chen
- Memory Ageing and Cognition Centre (MACC), National University Health System, Singapore.,Department of Pharmacology, National University of Singapore, Singapore
| | - Mohammad Kamran Ikram
- Memory Ageing and Cognition Centre (MACC), National University Health System, Singapore.,Department of Pharmacology, National University of Singapore, Singapore.,Singapore Eye Research Institute, Singapore National Eye Center, Singapore.,Academic Medicine Research Institute, Duke-NUS Graduate Medical School, Singapore
| |
Collapse
|
131
|
Zuloaga KL, Johnson LA, Roese NE, Marzulla T, Zhang W, Nie X, Alkayed FN, Hong C, Grafe MR, Pike MM, Raber J, Alkayed NJ. High fat diet-induced diabetes in mice exacerbates cognitive deficit due to chronic hypoperfusion. J Cereb Blood Flow Metab 2016; 36:1257-70. [PMID: 26661233 PMCID: PMC4929700 DOI: 10.1177/0271678x15616400] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/30/2015] [Indexed: 11/16/2022]
Abstract
Diabetes causes endothelial dysfunction and increases the risk of vascular cognitive impairment. However, it is unknown whether diabetes causes cognitive impairment due to reductions in cerebral blood flow or through independent effects on neuronal function and cognition. We addressed this using right unilateral common carotid artery occlusion to model vascular cognitive impairment and long-term high-fat diet to model type 2 diabetes in mice. Cognition was assessed using novel object recognition task, Morris water maze, and contextual and cued fear conditioning. Cerebral blood flow was assessed using arterial spin labeling magnetic resonance imaging. Vascular cognitive impairment mice showed cognitive deficit in the novel object recognition task, decreased cerebral blood flow in the right hemisphere, and increased glial activation in white matter and hippocampus. Mice fed a high-fat diet displayed deficits in the novel object recognition task, Morris water maze and fear conditioning tasks and neuronal loss, but no impairments in cerebral blood flow. Compared to vascular cognitive impairment mice fed a low fat diet, vascular cognitive impairment mice fed a high-fat diet exhibited reduced cued fear memory, increased deficit in the Morris water maze, neuronal loss, glial activation, and global decrease in cerebral blood flow. We conclude that high-fat diet and chronic hypoperfusion impair cognitive function by different mechanisms, although they share commons features, and that high-fat diet exacerbates vascular cognitive impairment pathology.
Collapse
Affiliation(s)
- Kristen L Zuloaga
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Lance A Johnson
- The Knight Cardiovascular Institute, Portland, OR, USA Department of Behavioral Neuroscience, Portland, OR, USA
| | - Natalie E Roese
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Tessa Marzulla
- Department of Behavioral Neuroscience, Portland, OR, USA
| | - Wenri Zhang
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Xiao Nie
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Farah N Alkayed
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Christine Hong
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Marjorie R Grafe
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR, USA Department of Pathology, Portland, OR, USA
| | - Martin M Pike
- Advanced Imaging Resource Center, Oregon Health and Science University, Portland, OR, USA
| | - Jacob Raber
- The Knight Cardiovascular Institute, Portland, OR, USA Department of Behavioral Neuroscience, Portland, OR, USA Departments of Neurology and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR, USA
| | - Nabil J Alkayed
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR, USA The Knight Cardiovascular Institute, Portland, OR, USA
| |
Collapse
|
132
|
YOO DY, YIM HS, JUNG HY, NAM SM, KIM JW, CHOI JH, SEONG JK, YOON YS, KIM DW, HWANG IK. Chronic type 2 diabetes reduces the integrity of the blood-brain barrier by reducing tight junction proteins in the hippocampus. J Vet Med Sci 2016; 78:957-62. [PMID: 26876499 PMCID: PMC4937155 DOI: 10.1292/jvms.15-0589] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 02/04/2016] [Indexed: 12/11/2022] Open
Abstract
In the present study, we investigated the effects of type 2 diabetes-induced hyperglycemia on the integrity of the blood-brain barrier and tight junction markers in the rat hippocampus. Forty-week-old diabetic (Zucker diabetic fatty, ZDF) rats and littermate control (Zucker lean control, ZLC) rats were used in this study. We evaluated the integrity of the blood-brain barrier by measuring sodium fluorescein extravasation and blood vessel ultrastructure. In addition, tight junction markers, such as zona occludens-1, occludin and claudin-5, were quantified by western blot analysis. ZDF rats showed significantly increased sodium fluorescein leakage in the hippocampus. Tight junction markers, such as occludin and claudin-5, were significantly decreased in the hippocampi of ZDF rats compared to those of ZLC rats. In addition, ZDF rats showed ultrastructural changes with phagocytic findings in the blood vessels. These results suggest that chronic untreated diabetes impairs the permeability of the hippocampal blood-brain barrier by down-regulating occludin and claudin-5, indicating that chronic untreated diabetes may cause hippocampus-dependent dysfunction.
Collapse
Affiliation(s)
- Dae Young YOO
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and
Research Institute for Veterinary Science, Seoul National University, Seoul 08826, South Korea
| | - Hee Sun YIM
- Department of Biochemistry and Molecular Biology, Research Institute of Oral
Sciences, College of Dentistry, Kangneung-Wonju National University, Gangneung 25457; South Korea
| | - Hyo Young JUNG
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and
Research Institute for Veterinary Science, Seoul National University, Seoul 08826, South Korea
| | - Sung Min NAM
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and
Research Institute for Veterinary Science, Seoul National University, Seoul 08826, South Korea
| | - Jong Whi KIM
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and
Research Institute for Veterinary Science, Seoul National University, Seoul 08826, South Korea
| | - Jung Hoon CHOI
- Department of Anatomy, College of Veterinary Medicine, Kangwon National
University, Chuncheon 24341, South Korea
| | - Je Kyung SEONG
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and
Research Institute for Veterinary Science, Seoul National University, Seoul 08826, South Korea
- KMPC (Korea Mouse Phenotyping Center), Seoul National University, Seoul 08826,
South Korea
| | - Yeo Sung YOON
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and
Research Institute for Veterinary Science, Seoul National University, Seoul 08826, South Korea
- KMPC (Korea Mouse Phenotyping Center), Seoul National University, Seoul 08826,
South Korea
| | - Dae Won KIM
- Department of Biochemistry and Molecular Biology, Research Institute of Oral
Sciences, College of Dentistry, Kangneung-Wonju National University, Gangneung 25457; South Korea
| | - In Koo HWANG
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and
Research Institute for Veterinary Science, Seoul National University, Seoul 08826, South Korea
- KMPC (Korea Mouse Phenotyping Center), Seoul National University, Seoul 08826,
South Korea
| |
Collapse
|
133
|
Cui Y, Li SF, Gu H, Hu YZ, Liang X, Lu CQ, Cai Y, Wang CX, Yang Y, Teng GJ. Disrupted Brain Connectivity Patterns in Patients with Type 2 Diabetes. AJNR Am J Neuroradiol 2016; 37:2115-2122. [PMID: 27365332 DOI: 10.3174/ajnr.a4858] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 05/11/2016] [Indexed: 01/12/2023]
Abstract
BACKGROUND AND PURPOSE Type 2 diabetes is associated with an increased risk of dementia. This study investigated the global connectivity patterns in the brains of patients with type 2 diabetes by using a functional MR imaging technique. MATERIALS AND METHODS Forty patients and 43 age-, sex-, and education-matched healthy controls underwent resting-state functional imaging in a 3T MR imaging unit. Degree centrality, a commonly used measurement of global connectivity, was computed for a full-brain exploration of the regions influenced by type 2 diabetes. We then examined the functional connectivity of each region by using the seed-based approach. Finally, voxelwise correlation analyses were performed to explore the relationship among the connectivity changes, cognitive performance, and diabetes-related variables. RESULTS Patients exhibited decreased degree centrality in the left lingual gyrus and increased centrality in the right insula and dorsal anterior cingulate cortex (corrected P < .05). The occipital network anchored in the lingual gyrus showed extensively reduced connectivity, while the network connectivity of the insula and cingulate cortex (mostly included in the salience network) was significantly elevated (corrected P < .05). Correlational analyses revealed that in the diabetic group, impaired visual memory and executive function performance were correlated with occipital hypoconnectivity, while higher fasting plasma glucose levels and better executive functioning were related to anterior cingulate cortex hyperconnectivity (all corrected P values < .05). Similar effects were not detected in the controls. CONCLUSIONS This preliminary study shows that network connectivity is altered in patients with type 2 diabetes, which may provide critical insight into the neural substrate of diabetes-related cognitive decline.
Collapse
Affiliation(s)
- Y Cui
- From the Department of Radiology (Y.Cui, C.-Q.L., Y.Cai, C-X.W., G.-J.T.), Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, Medical School of Southeast University, Nanjing, China.,Neuroimaging Research Branch (Y.Cui, S.-F.L., H.G., Y.-Z.H, X.L., Y.Y.), National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland
| | - S-F Li
- Neuroimaging Research Branch (Y.Cui, S.-F.L., H.G., Y.-Z.H, X.L., Y.Y.), National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland
| | - H Gu
- Neuroimaging Research Branch (Y.Cui, S.-F.L., H.G., Y.-Z.H, X.L., Y.Y.), National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland
| | - Y-Z Hu
- Neuroimaging Research Branch (Y.Cui, S.-F.L., H.G., Y.-Z.H, X.L., Y.Y.), National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland
| | - X Liang
- Neuroimaging Research Branch (Y.Cui, S.-F.L., H.G., Y.-Z.H, X.L., Y.Y.), National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland
| | - C-Q Lu
- From the Department of Radiology (Y.Cui, C.-Q.L., Y.Cai, C-X.W., G.-J.T.), Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Y Cai
- From the Department of Radiology (Y.Cui, C.-Q.L., Y.Cai, C-X.W., G.-J.T.), Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - C-X Wang
- From the Department of Radiology (Y.Cui, C.-Q.L., Y.Cai, C-X.W., G.-J.T.), Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Y Yang
- Neuroimaging Research Branch (Y.Cui, S.-F.L., H.G., Y.-Z.H, X.L., Y.Y.), National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland
| | - G-J Teng
- From the Department of Radiology (Y.Cui, C.-Q.L., Y.Cai, C-X.W., G.-J.T.), Jiangsu Key Laboratory of Molecular and Functional Imaging, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| |
Collapse
|
134
|
Zhao Q, Roberts RO, Ding D, Cha R, Guo Q, Meng H, Luo J, Machulda MM, Shane Pankratz V, Wang B, Christianson TJH, Aakre JA, Knopman DS, Boeve BF, Hong Z, Petersen RC. Diabetes is Associated with Worse Executive Function in Both Eastern and Western Populations: Shanghai Aging Study and Mayo Clinic Study of Aging. J Alzheimers Dis 2016; 47:167-76. [PMID: 26402765 DOI: 10.3233/jad-150073] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND OBJECTIVES It remains unknown whether the association between diabetes mellitus (DM) and cognitive function differs in Eastern and Western populations. This study aimed to elucidate whether DM is associated with worse cognitive performance in both populations. METHODS The Shanghai Aging Study (SAS) and the Mayo Clinic Study of Aging (MCSA) are two population-based studies with similar design and methodology in Shanghai, China and Rochester, MN, USA. Non-demented participants underwent cognitive testing, and DM was assessed from the medical record. Separate analyses were performed in SAS and MCSA regarding the association between DM and cognitive performance. RESULTS A total of 3,348 Chinese participants in the SAS and 3,734 American subjects in the MCSA were included. Compared with MCSA subjects, SAS participants were younger, less educated, and had lower frequency of vascular disease, APOE ɛ4 carriers and obesity. Participants with DM (compared to non-DM participants) performed significantly worse on all the cognitive domains in both the SAS and MCSA. After adjustment for age, gender, education, and vascular covariates, DM was associated with worse performance in executive function (β=-0.15, p = 0.001 for SAS, and β=-0.10, p = 0.008 for MCSA) in the total sample and in the cognitively normal sub-sample. Furthermore, DM was associated with poor performance in visuospatial skills, language, and memory in the SAS, but not in the MCSA. CONCLUSIONS Diabetes is associated with cognitive dysfunction and, in particular, exerts a negative impact on executive function regardless of race, age, and prevalence of vascular risk factors.
Collapse
Affiliation(s)
- Qianhua Zhao
- Department of Neurology, Huashan Hospital, Fudan University, WHO Collaborating Center for Research and Training in Neurosciences, Shanghai, China
| | - Rosebud O Roberts
- Divisions of Epidemiology and Biomedical Statistics & Informatics, Mayo Clinic, Rochester, Minnesota, USA.,Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ding Ding
- Department of Neurology, Huashan Hospital, Fudan University, WHO Collaborating Center for Research and Training in Neurosciences, Shanghai, China
| | - Ruth Cha
- Divisions of Epidemiology and Biomedical Statistics & Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Qihao Guo
- Department of Neurology, Huashan Hospital, Fudan University, WHO Collaborating Center for Research and Training in Neurosciences, Shanghai, China
| | - Haijiao Meng
- Department of Neurology, Huashan Hospital, Fudan University, WHO Collaborating Center for Research and Training in Neurosciences, Shanghai, China
| | - Jianfeng Luo
- Department of Health Statistics, School of Public Health, Fudan University, Shanghai, China
| | - Mary M Machulda
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA
| | - V Shane Pankratz
- Divisions of Epidemiology and Biomedical Statistics & Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Bei Wang
- Department of Neurology, Huashan Hospital, Fudan University, WHO Collaborating Center for Research and Training in Neurosciences, Shanghai, China
| | - Teresa J H Christianson
- Divisions of Epidemiology and Biomedical Statistics & Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Jeremiah A Aakre
- Divisions of Epidemiology and Biomedical Statistics & Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - David S Knopman
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Bradley F Boeve
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Zhen Hong
- Department of Neurology, Huashan Hospital, Fudan University, WHO Collaborating Center for Research and Training in Neurosciences, Shanghai, China
| | | | | | | |
Collapse
|
135
|
Hou YC, Lai CH, Wu YT, Yang SH. Gray matter alterations and correlation of nutritional intake with the gray matter volume in prediabetes. Medicine (Baltimore) 2016; 95:e3956. [PMID: 27336893 PMCID: PMC4998331 DOI: 10.1097/md.0000000000003956] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The neurophysiology of prediabetes plays an important role in preventive medicine. The dysregulation of glucose metabolism is likely linked to changes in neuron-related gray matter. Therefore, we designed this study to investigate gray matter alterations in medication-naive prediabetic patients. We expected to find alterations in the gray matter of prediabetic patients.A total of 64 prediabetic patients and 54 controls were enrolled. All subjects received T1 scans using a 3-T magnetic resonance imaging machine. Subjects also completed nutritional intake records at the 24-hour and 3-day time points to determine their carbohydrate, protein, fat, and total calorie intake. We utilized optimized voxel-based morphometry to estimate the gray matter differences between the patients and controls. In addition, the preprandial serum glucose level and the carbohydrate, protein, fat, and total calorie intake levels were tested to determine whether these parameters were correlated with the gray matter volume.Prediabetic patients had lower gray matter volumes than controls in the right anterior cingulate gyrus, right posterior cingulate gyrus, left insula, left super temporal gyrus, and left middle temporal gyrus (corrected P < 0.05; voxel threshold: 33). Gray matter volume in the right anterior cingulate was also negatively correlated with the preprandial serum glucose level gyrus in a voxel-dependent manner (r = -0.501; 2-tailed P = 0.001).The cingulo-temporal and insula gray matter alterations may be associated with the glucose dysregulation in prediabetic patients.
Collapse
Affiliation(s)
- Yi-Cheng Hou
- Department of Nutrition, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City
- School of Nutrition and Health Sciences, College of Public Health and Nutrition, Taipei Medical University
| | - Chien-Han Lai
- Department of Biomedical Imaging and Radiological Sciences
- Institute of Biophotonics, National Yang-Ming University, Taipei
- Department of Psychiatry, Cheng Hsin General Hospital, Taipei City, Taiwan, ROC
| | - Yu-Te Wu
- Department of Biomedical Imaging and Radiological Sciences
- Institute of Biophotonics, National Yang-Ming University, Taipei
| | - Shwu-Huey Yang
- School of Nutrition and Health Sciences, College of Public Health and Nutrition, Taipei Medical University
| |
Collapse
|
136
|
Diabetes and Alzheimer’s disease crosstalk. Neurosci Biobehav Rev 2016; 64:272-87. [PMID: 26969101 DOI: 10.1016/j.neubiorev.2016.03.005] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 01/26/2016] [Accepted: 03/04/2016] [Indexed: 12/12/2022]
|
137
|
Vassilaki M, Aakre JA, Mielke MM, Geda YE, Kremers WK, Alhurani RE, Machulda MM, Knopman DS, Petersen RC, Lowe VJ, Jack CR, Roberts RO. Multimorbidity and neuroimaging biomarkers among cognitively normal persons. Neurology 2016; 86:2077-84. [PMID: 27164657 DOI: 10.1212/wnl.0000000000002624] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 02/24/2016] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To assess the cross-sectional association between multimorbidity and imaging biomarkers of brain pathology in the population-based Mayo Clinic Study of Aging (MCSA). METHODS The study consisted of 1,449 MCSA participants who were cognitively normal at the time of MRI. A subset of the participants also had (11)C-Pittsburgh compound B (n = 689) and (18)fluorodeoxyglucose (n = 688) PET scans available. Information on multimorbidity (defined as ≥2 chronic conditions) in the 5 years prior to the first imaging study was captured from the medical record using ICD-9 codes for chronic conditions and the Rochester Epidemiology Project medical records linkage system. The cross-sectional association of multimorbidity and imaging biomarkers was examined using logistic and linear regression models. RESULTS Among 1,449 cognitively normal participants (mean age 79 years; 50.9% men), 85.4% had multimorbidity (≥2 chronic conditions). Multimorbidity and severe multimorbidity (≥4 chronic conditions) were associated with abnormal Alzheimer disease (AD) signature meta-region of interest (meta-ROI) (18)F-FDG hypometabolism (odds ratio [OR] 2.03; 95% confidence interval [CI] 1.10-3.77 and OR 2.22; 95% CI 1.18-4.16, respectively), and with abnormal AD signature MRI cortical thickness (OR 1.53; 95% CI 1.09-2.16 and OR 1.76; 95% CI 1.24-2.51, respectively), but was not associated with amyloid accumulation. CONCLUSIONS Multimorbidity was associated with brain pathology through mechanisms independent of amyloid deposition and such neuronal injury and pathology was present before any symptomatic evidence of cognitive impairment. Longitudinal follow-up will provide insights into potential causal associations of multimorbidity with changes in brain pathology.
Collapse
Affiliation(s)
- Maria Vassilaki
- From the Departments of Health Sciences Research (M.V., J.A.A., M.M. Mielke, Y.E.G., W.K.K., R.C.P., R.O.R.) and Neurology (M.M. Mielke, Y.E.G., R.E.A., D.S.K., R.C.P., R.O.R.), Mayo Clinic, Rochester, MN; Departments of Psychiatry and Psychology and Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ; Mayo Clinic Graduate School of Medicine (R.E.A.), Rochester, MN; and Departments of Psychiatry and Psychology (M.M. Machulda) and Radiology (V.J.L., C.R.J.), Mayo Clinic, Rochester, MN.
| | - Jeremiah A Aakre
- From the Departments of Health Sciences Research (M.V., J.A.A., M.M. Mielke, Y.E.G., W.K.K., R.C.P., R.O.R.) and Neurology (M.M. Mielke, Y.E.G., R.E.A., D.S.K., R.C.P., R.O.R.), Mayo Clinic, Rochester, MN; Departments of Psychiatry and Psychology and Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ; Mayo Clinic Graduate School of Medicine (R.E.A.), Rochester, MN; and Departments of Psychiatry and Psychology (M.M. Machulda) and Radiology (V.J.L., C.R.J.), Mayo Clinic, Rochester, MN
| | - Michelle M Mielke
- From the Departments of Health Sciences Research (M.V., J.A.A., M.M. Mielke, Y.E.G., W.K.K., R.C.P., R.O.R.) and Neurology (M.M. Mielke, Y.E.G., R.E.A., D.S.K., R.C.P., R.O.R.), Mayo Clinic, Rochester, MN; Departments of Psychiatry and Psychology and Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ; Mayo Clinic Graduate School of Medicine (R.E.A.), Rochester, MN; and Departments of Psychiatry and Psychology (M.M. Machulda) and Radiology (V.J.L., C.R.J.), Mayo Clinic, Rochester, MN
| | - Yonas E Geda
- From the Departments of Health Sciences Research (M.V., J.A.A., M.M. Mielke, Y.E.G., W.K.K., R.C.P., R.O.R.) and Neurology (M.M. Mielke, Y.E.G., R.E.A., D.S.K., R.C.P., R.O.R.), Mayo Clinic, Rochester, MN; Departments of Psychiatry and Psychology and Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ; Mayo Clinic Graduate School of Medicine (R.E.A.), Rochester, MN; and Departments of Psychiatry and Psychology (M.M. Machulda) and Radiology (V.J.L., C.R.J.), Mayo Clinic, Rochester, MN
| | - Walter K Kremers
- From the Departments of Health Sciences Research (M.V., J.A.A., M.M. Mielke, Y.E.G., W.K.K., R.C.P., R.O.R.) and Neurology (M.M. Mielke, Y.E.G., R.E.A., D.S.K., R.C.P., R.O.R.), Mayo Clinic, Rochester, MN; Departments of Psychiatry and Psychology and Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ; Mayo Clinic Graduate School of Medicine (R.E.A.), Rochester, MN; and Departments of Psychiatry and Psychology (M.M. Machulda) and Radiology (V.J.L., C.R.J.), Mayo Clinic, Rochester, MN
| | - Rabe E Alhurani
- From the Departments of Health Sciences Research (M.V., J.A.A., M.M. Mielke, Y.E.G., W.K.K., R.C.P., R.O.R.) and Neurology (M.M. Mielke, Y.E.G., R.E.A., D.S.K., R.C.P., R.O.R.), Mayo Clinic, Rochester, MN; Departments of Psychiatry and Psychology and Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ; Mayo Clinic Graduate School of Medicine (R.E.A.), Rochester, MN; and Departments of Psychiatry and Psychology (M.M. Machulda) and Radiology (V.J.L., C.R.J.), Mayo Clinic, Rochester, MN
| | - Mary M Machulda
- From the Departments of Health Sciences Research (M.V., J.A.A., M.M. Mielke, Y.E.G., W.K.K., R.C.P., R.O.R.) and Neurology (M.M. Mielke, Y.E.G., R.E.A., D.S.K., R.C.P., R.O.R.), Mayo Clinic, Rochester, MN; Departments of Psychiatry and Psychology and Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ; Mayo Clinic Graduate School of Medicine (R.E.A.), Rochester, MN; and Departments of Psychiatry and Psychology (M.M. Machulda) and Radiology (V.J.L., C.R.J.), Mayo Clinic, Rochester, MN
| | - David S Knopman
- From the Departments of Health Sciences Research (M.V., J.A.A., M.M. Mielke, Y.E.G., W.K.K., R.C.P., R.O.R.) and Neurology (M.M. Mielke, Y.E.G., R.E.A., D.S.K., R.C.P., R.O.R.), Mayo Clinic, Rochester, MN; Departments of Psychiatry and Psychology and Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ; Mayo Clinic Graduate School of Medicine (R.E.A.), Rochester, MN; and Departments of Psychiatry and Psychology (M.M. Machulda) and Radiology (V.J.L., C.R.J.), Mayo Clinic, Rochester, MN
| | - Ronald C Petersen
- From the Departments of Health Sciences Research (M.V., J.A.A., M.M. Mielke, Y.E.G., W.K.K., R.C.P., R.O.R.) and Neurology (M.M. Mielke, Y.E.G., R.E.A., D.S.K., R.C.P., R.O.R.), Mayo Clinic, Rochester, MN; Departments of Psychiatry and Psychology and Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ; Mayo Clinic Graduate School of Medicine (R.E.A.), Rochester, MN; and Departments of Psychiatry and Psychology (M.M. Machulda) and Radiology (V.J.L., C.R.J.), Mayo Clinic, Rochester, MN
| | - Val J Lowe
- From the Departments of Health Sciences Research (M.V., J.A.A., M.M. Mielke, Y.E.G., W.K.K., R.C.P., R.O.R.) and Neurology (M.M. Mielke, Y.E.G., R.E.A., D.S.K., R.C.P., R.O.R.), Mayo Clinic, Rochester, MN; Departments of Psychiatry and Psychology and Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ; Mayo Clinic Graduate School of Medicine (R.E.A.), Rochester, MN; and Departments of Psychiatry and Psychology (M.M. Machulda) and Radiology (V.J.L., C.R.J.), Mayo Clinic, Rochester, MN
| | - Clifford R Jack
- From the Departments of Health Sciences Research (M.V., J.A.A., M.M. Mielke, Y.E.G., W.K.K., R.C.P., R.O.R.) and Neurology (M.M. Mielke, Y.E.G., R.E.A., D.S.K., R.C.P., R.O.R.), Mayo Clinic, Rochester, MN; Departments of Psychiatry and Psychology and Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ; Mayo Clinic Graduate School of Medicine (R.E.A.), Rochester, MN; and Departments of Psychiatry and Psychology (M.M. Machulda) and Radiology (V.J.L., C.R.J.), Mayo Clinic, Rochester, MN
| | - Rosebud O Roberts
- From the Departments of Health Sciences Research (M.V., J.A.A., M.M. Mielke, Y.E.G., W.K.K., R.C.P., R.O.R.) and Neurology (M.M. Mielke, Y.E.G., R.E.A., D.S.K., R.C.P., R.O.R.), Mayo Clinic, Rochester, MN; Departments of Psychiatry and Psychology and Neurology (Y.E.G.), Mayo Clinic, Scottsdale, AZ; Mayo Clinic Graduate School of Medicine (R.E.A.), Rochester, MN; and Departments of Psychiatry and Psychology (M.M. Machulda) and Radiology (V.J.L., C.R.J.), Mayo Clinic, Rochester, MN.
| |
Collapse
|
138
|
Winblad B, Amouyel P, Andrieu S, Ballard C, Brayne C, Brodaty H, Cedazo-Minguez A, Dubois B, Edvardsson D, Feldman H, Fratiglioni L, Frisoni GB, Gauthier S, Georges J, Graff C, Iqbal K, Jessen F, Johansson G, Jönsson L, Kivipelto M, Knapp M, Mangialasche F, Melis R, Nordberg A, Rikkert MO, Qiu C, Sakmar TP, Scheltens P, Schneider LS, Sperling R, Tjernberg LO, Waldemar G, Wimo A, Zetterberg H. Defeating Alzheimer's disease and other dementias: a priority for European science and society. Lancet Neurol 2016; 15:455-532. [DOI: 10.1016/s1474-4422(16)00062-4] [Citation(s) in RCA: 1001] [Impact Index Per Article: 111.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 10/06/2015] [Accepted: 02/09/2016] [Indexed: 12/15/2022]
|
139
|
Rodríguez-Sánchez E, Mora-Simón S, Patino-Alonso MC, Pérez-Arechaederra D, Recio-Rodríguez JI, Gómez-Marcos MA, Valero-Juan LF, García-Ortiz L. Cognitive impairment and dependence of patients with diabetes older than 65 years old in an urban area (DERIVA study). BMC Geriatr 2016; 16:33. [PMID: 26832143 PMCID: PMC4736631 DOI: 10.1186/s12877-016-0208-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 01/26/2016] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND We analyzed the associations between diabetes and cognitive impairment (CI) and dependence in a population of patients 65 years or older. METHODS Cross-sectional study. We randomly selected 311 participants over the age of 65 living in an urban area of Spain. The mean age of the cohort was 75.89 ± 7.12 years, and 69 of the individuals (22.2%) had diabetes. Two questionnaires were used to assess cognitive performance (MMSE and Seven Minute Screen Test), and two assessments were used to evaluate patient dependence (Barthel Index and Lawton-Brody Index). Clinical information and sociodemographic data were also gathered. RESULTS Nearly one quarter of patients with diabetes (21.7%) lived alone. Diabetic patients were more sedentary (p = .033) than non-diabetic patients. Roughly one sixth (15.3%) of the diabetics and 10.1% of the non-diabetics were depressed (p = .332). CI was present in 26.1% of the diabetics and 14.5% of non-diabetics (p = .029). Diabetic patients had a MMSE score that was significantly worse than non-diabetics (24.88 ± 4.74 vs 26.05 ± 4.03; p <.05), but no differences were found in the Seven Minute Screen Test. Logistic regressions revealed that the presence of diabetes was independently associated with CI (adjusted for age, gender, years of education, sedentary lifestyle, body mass index, diastolic blood pressure, cholesterol, and depression (OR = 2.940, p = .013). Patients with diabetes showed greater dependence, as measured by the Barthel Index (p = .03) and Lawton-Brody Index (p <.01). Nevertheless, when dependence (dependence or not dependence for each questionnaire) used as a dependent variable in the logistic regression analyses, no significant association with diabetes was found, after adjusting for confounding variables. CONCLUSIONS Diabetic patients over the age of 65 are more likely to present CI but not dependence. These findings support the need to include both a functional and cognitive assessment as necessary components in a standard evaluation in both clinical guides and randomized trials of therapeutic interventions in patients with diabetes.
Collapse
Affiliation(s)
- Emiliano Rodríguez-Sánchez
- Primary Care Research Unit, The Alamedilla Health Center, Castilla and León Health Service (SACyL), Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain. .,Medicine Department, University of Salamanca, Salamanca, Spain.
| | - Sara Mora-Simón
- Primary Care Research Unit, The Alamedilla Health Center, Castilla and León Health Service (SACyL), Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain. .,Basic Psychology, Psychobiology and Behavioral Sciences Methodology Department, University of Salamanca, Salamanca, Spain.
| | - María C Patino-Alonso
- Primary Care Research Unit, The Alamedilla Health Center, Castilla and León Health Service (SACyL), Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain. .,Statistics Department, University of Salamanca, Salamanca, Spain.
| | - Diana Pérez-Arechaederra
- Primary Care Research Unit, The Alamedilla Health Center, Castilla and León Health Service (SACyL), Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain.
| | - José I Recio-Rodríguez
- Primary Care Research Unit, The Alamedilla Health Center, Castilla and León Health Service (SACyL), Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain.
| | - Manuel A Gómez-Marcos
- Primary Care Research Unit, The Alamedilla Health Center, Castilla and León Health Service (SACyL), Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain. .,Medicine Department, University of Salamanca, Salamanca, Spain.
| | - Luis F Valero-Juan
- Primary Care Research Unit, The Alamedilla Health Center, Castilla and León Health Service (SACyL), Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain. .,Preventive Medicine, Public Health and Medical Microbiology Department, University of Salamanca, Salamanca, Spain.
| | - Luis García-Ortiz
- Primary Care Research Unit, The Alamedilla Health Center, Castilla and León Health Service (SACyL), Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain. .,Biomedical and Diagnostic Sciences Department, University of Salamanca, Salamanca, Spain.
| | | |
Collapse
|
140
|
Rettberg JR, Dang H, Hodis HN, Henderson VW, St John JA, Mack WJ, Brinton RD. Identifying postmenopausal women at risk for cognitive decline within a healthy cohort using a panel of clinical metabolic indicators: potential for detecting an at-Alzheimer's risk metabolic phenotype. Neurobiol Aging 2016; 40:155-163. [PMID: 26973115 DOI: 10.1016/j.neurobiolaging.2016.01.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 01/08/2016] [Accepted: 01/21/2016] [Indexed: 12/20/2022]
Abstract
Detecting at-risk individuals within a healthy population is critical for preventing or delaying Alzheimer's disease. Systems biology integration of brain and body metabolism enables peripheral metabolic biomarkers to serve as reporters of brain bioenergetic status. Using clinical metabolic data derived from healthy postmenopausal women in the Early versus Late Intervention Trial with Estradiol (ELITE), we conducted principal components and k-means clustering analyses of 9 biomarkers to define metabolic phenotypes. Metabolic clusters were correlated with cognitive performance and analyzed for change over 5 years. Metabolic biomarkers at baseline generated 3 clusters, representing women with healthy, high blood pressure, and poor metabolic phenotypes. Compared with healthy women, poor metabolic women had significantly lower executive, global and memory cognitive performance. Hormone therapy provided metabolic benefit to women in high blood pressure and poor metabolic phenotypes. This panel of well-established clinical peripheral biomarkers represents an initial step toward developing an affordable, rapidly deployable, and clinically relevant strategy to detect an at-risk phenotype of late-onset Alzheimer's disease.
Collapse
Affiliation(s)
- Jamaica R Rettberg
- Neuroscience Department, University of Southern California, Los Angeles, CA, USA
| | - Ha Dang
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Howard N Hodis
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA; Department of Medicine, Atherosclerosis Research Unit, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Victor W Henderson
- Division of Epidemiology, Department of Health Research and Policy, Stanford University, Stanford, CA, USA; Department of Neurology & Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Jan A St John
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA; Department of Medicine, Atherosclerosis Research Unit, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Wendy J Mack
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA; Department of Medicine, Atherosclerosis Research Unit, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Roberta Diaz Brinton
- Neuroscience Department, University of Southern California, Los Angeles, CA, USA; Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA; Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
141
|
Spitznagel MB, Alosco M, Inge TH, Rochette A, Strain G, Devlin M, Crosby RD, Mitchell JE, Gunstad J. Adolescent weight history and adult cognition: before and after bariatric surgery. Surg Obes Relat Dis 2016; 12:1057-1064. [PMID: 27178608 DOI: 10.1016/j.soard.2016.01.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 01/26/2016] [Accepted: 01/26/2016] [Indexed: 10/22/2022]
Abstract
BACKGROUND Cognitive deficits occur in a subset of individuals with obesity. Deficits can be reversed with bariatric surgery, though cognitive recovery is not equally exhibited across patients. Recent work has found that obesity during adolescence portends medical complications in adulthood; it is unknown if obesity in adolescence predicts adult cognition or cognitive recovery after weight loss surgery. OBJECTIVES The present study examines the relationship between weight history and cognitive function in obese adults undergoing bariatric surgery. SETTING Academic medical centers with bariatric care services. METHODS Seventy-eight bariatric surgery patients (mean age = 43.2 years) enrolled in an ancillary study to the Longitudinal Assessment of Bariatric Surgery (LABS) project completed a questionnaire recalling weight history at age 18. Cognitive testing was completed preoperatively and at 12-month follow-up. RESULTS Weight status at age 18 was linked to performance in several aspects of cognition. Higher body mass index at age 18 predicted poorer preoperative verbal fluency (B = -.26, P = .045) as well as postoperative cognitive recovery in attention (B = -.30, P = .01) at 12-month follow-up. CONCLUSION Higher body mass index at age 18 predicts verbal fluency performance in adults with obesity, as well as postoperative recovery of attention after bariatric surgery. The mechanisms underlying this connection are not fully clear, though findings may reflect effects of obesity on the brain during a crucial period of neural maturation or duration of obesity and cumulative impact of co-morbidities on cognition. Future work examining possible causal factors involved in these relationships is needed.
Collapse
Affiliation(s)
| | | | - Thomas H Inge
- Cincinnati Children's Hospital Medical Center and University of Cincinnati, Cincinnati, Ohio
| | | | | | - Michael Devlin
- Columbia University College of Physicians & Surgeons, New York, New York
| | - Ross D Crosby
- Neuropsychiatric Research Institute and University of North Dakota School of Medicine and Health Sciences, Fargo, North Dakota
| | - James E Mitchell
- Neuropsychiatric Research Institute and University of North Dakota School of Medicine and Health Sciences, Fargo, North Dakota
| | - John Gunstad
- Kent State University, Kent, Ohio; Summa Health System, Akron, Ohio
| |
Collapse
|
142
|
Drewes AM, Søfteland E, Dimcevski G, Farmer AD, Brock C, Frøkjær JB, Krogh K, Drewes AM. Brain changes in diabetes mellitus patients with gastrointestinal symptoms. World J Diabetes 2016; 7:14-26. [PMID: 26839652 PMCID: PMC4724575 DOI: 10.4239/wjd.v7.i2.14] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 09/14/2015] [Accepted: 10/27/2015] [Indexed: 02/05/2023] Open
Abstract
Diabetes mellitus is a common disease and its prevalence is increasing worldwide. In various studies up to 30%-70% of patients present dysfunction and complications related to the gut. To date several clinical studies have demonstrated that autonomic nervous system neuropathy and generalized neuropathy of the central nervous system (CNS) may play a major role. This systematic review provides an overview of the neurodegenerative changes that occur as a consequence of diabetes with a focus on the CNS changes and gastrointestinal (GI) dysfunction. Animal models where diabetes was induced experimentally support that the disease induces changes in CNS. Recent investigations with electroencephalography and functional brain imaging in patients with diabetes confirm these structural and functional brain changes. Encephalographic studies demonstrated that altered insular processing of sensory stimuli seems to be a key player in symptom generation. In fact one study indicated that the more GI symptoms the patients experienced, the deeper the insular electrical source was located. The electroencephalography was often used in combination with quantitative sensory testing mainly showing hyposensitivity to stimulation of GI organs. Imaging studies on patients with diabetes and GI symptoms mainly showed microstructural changes, especially in brain areas involved in visceral sensory processing. As the electrophysiological and imaging changes were associated with GI and autonomic symptoms they may represent a future therapeutic target for treating diabetics either pharmacologically or with neuromodulation.
Collapse
|
143
|
The neurobiology of HIV and its impact on cognitive reserve: A review of cognitive interventions for an aging population. Neurobiol Dis 2016; 92:144-56. [PMID: 26776767 DOI: 10.1016/j.nbd.2016.01.011] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 12/17/2015] [Accepted: 01/13/2016] [Indexed: 12/21/2022] Open
Abstract
The medications used to treat HIV have reduced the severity of cognitive deficits; yet, nearly half of adults with HIV still exhibit some degree of cognitive deficits, referred to as HIV-associated neurocognitive disorder or HAND. These cognitive deficits interfere with everyday functioning such as emotional regulation, medication adherence, instrumental activities of daily living, and even driving a vehicle. As adults are expected to live a normal lifespan, the process of aging in this clinical population may exacerbate such cognitive deficits. Therefore, it is important to understand the neurobiological mechanisms of HIV on cognitive reserve and develop interventions that are either neuroprotective or compensate for such cognitive deficits. Within the context of cognitive reserve, this article delivers a state of the science perspective on the causes of HAND and provides possible interventions for addressing such cognitive deficits. Suggestions for future research are also provided.
Collapse
|
144
|
Miwa K, Okazaki S, Sakaguchi M, Mochizuki H, Kitagawa K. Interleukin-6, interleukin-6 receptor gene variant, small-vessel disease and incident dementia. Eur J Neurol 2016; 23:656-63. [PMID: 26725994 DOI: 10.1111/ene.12921] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 11/04/2015] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND PURPOSE Mixed neurogenerative and vascular dementia has emerged as the leading cause of dementia in the elderly. Inflammation is implicated in atherosclerosis, cerebral small-vessel disease (SVD) as well as cognitive impairment. However, longitudinal data on the predictive value of circulating inflammatory markers including gene variants and magnetic resonance imaging (MRI) findings in incident dementia are scarce. It was investigated whether circulating interleukin-6 (IL-6), C-reactive protein (CRP) and gene variants increase dementia risk. METHODS In a cohort of Japanese participants with vascular risk factors in an observational study from 2001, the association between baseline IL-6, CRP levels, gene variants [interleukin-6 receptor (IL-6R), rs2228145; IL-6, rs2097677; CRP, rs3093059] and incident all-cause dementia was evaluated. Baseline MRI was used to determine SVD (lacuna, white matter hyperintensities) and atrophy (medial-temporal lobe atrophy, bicaudate ratio). Cox proportional hazards analyses were performed for predictors of dementia, adjusting for age, sex, apolipoprotein Eε4, education, cerebrovascular events, vascular risk factors and MRI findings. RESULTS Of 803 subjects (mean 67.0 ± 8.5 years, males 59%), during a mean of 7.5 ± 3.2 years follow-up, 60 incident dementia patients (Alzheimer's disease 31; vascular dementia 17; mixed-type six; other six) were diagnosed. In multivariable analyses adjusted for age, sex, cerebrovascular events, MRI findings and IL-6R variant (rs2228145), IL-6 levels (relative risk 1.68, P = 0.048) or highest tertile (relative risk 2.38, P = 0.031) for all-cause dementia remained significant. Although subjects with rs2228145 carrier had significantly higher IL-6 levels, a significant association between rs2228145 and dementia was not observed. Conversely, CRP and remaining gene variants were not associated with dementia. CONCLUSIONS The deleterious effect of higher IL-6 on dementia remains consistent irrespective of conventional risk factors, MRI findings and IL-6R variant.
Collapse
Affiliation(s)
- K Miwa
- Department of Neurology and Stroke Center, Osaka University Graduate School of Medicine, Osaka, Japan
| | - S Okazaki
- Department of Neurology and Stroke Center, Osaka University Graduate School of Medicine, Osaka, Japan
| | - M Sakaguchi
- Department of Neurology and Stroke Center, Osaka University Graduate School of Medicine, Osaka, Japan
| | - H Mochizuki
- Department of Neurology and Stroke Center, Osaka University Graduate School of Medicine, Osaka, Japan
| | - K Kitagawa
- Department of Neurology, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
145
|
|
146
|
Wang H, Chen F, Zhong KL, Tang SS, Hu M, Long Y, Miao MX, Liao JM, Sun HB, Hong H. PPARγ agonists regulate bidirectional transport of amyloid-β across the blood-brain barrier and hippocampus plasticity in db/db mice. Br J Pharmacol 2015; 173:372-85. [PMID: 26507867 DOI: 10.1111/bph.13378] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 10/18/2015] [Accepted: 10/21/2015] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND AND PURPOSE There is emerging evidence suggesting that abnormal transport of amyloid-β (Aβ) across the blood-brain barrier (BBB) is involved in diabetes-associated cognitive decline. We investigated whether PPARγ agonists restore Aβ transport across the BBB and hippocampal plasticity in db/db mice. EXPERIMENTAL APPROACH Efflux and influx of Aβ across the BBB were determined by stereotaxic intra-cerebral or i.a. infusion of [(125) I]-Aβ1-40 respectively. Receptor for advanced glycation end products (RAGE) and low-density lipoprotein receptor-related protein 1 (LRP1), which are involved in Aβ influx and efflux, PPARγ and NF-κB p65 at the BBB, as well as hippocampal Aβ, caspase-3, Bax and Bcl-2 were assayed by Western blot, immunohistochemistry and RT-PCR. In vivo, hippocampal LTP was recorded, and Morris water maze and Y-maze tasks were performed. KEY RESULTS Treatment with PPARγ agonists, rosiglitazone (0.8 mg·kg(-1) ) and pioglitazone (9.0 mg·kg(-1) ), for 6 weeks significantly increased Aβ efflux and decreased Aβ influx across the BBB in db/db mice. Concomitantly, they decreased hippocampal Aβ1-40 and Aβ1-42 , suppressed neuronal apoptosis, as indicated by decreased caspase-3 activity and increased ratio of Bcl-2/Bax, and increased hippocampal plasticity, characterized by an enhanced in vivo LTP and better performance in behavioural tests. Furthermore, the PPARγ agonists induced the expression of LRP1 gene by activation of PPARγ and suppressed RAGE gene expression by inactivation of NF-κB signalling at the BBB of db/db mice. CONCLUSIONS AND IMPLICATIONS PPARγ agonists modify abnormal Aβ transport across the BBB and this is accompanied by amelioration of β-amyloidosis and an improvement in hippocampal plasticity in diabetic mice.
Collapse
Affiliation(s)
- Hao Wang
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| | - Fang Chen
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| | - Kai Long Zhong
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| | - Su Su Tang
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| | - Mei Hu
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| | - Yan Long
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| | - Ming Xing Miao
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| | - Jian Min Liao
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| | - Hong Bing Sun
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| | - Hao Hong
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
147
|
Tsuruya K, Yoshida H, Haruyama N, Fujisaki K, Hirakata H, Kitazono T. Clinical Significance of Fronto-Temporal Gray Matter Atrophy in Executive Dysfunction in Patients with Chronic Kidney Disease: The VCOHP Study. PLoS One 2015; 10:e0143706. [PMID: 26632813 PMCID: PMC4669129 DOI: 10.1371/journal.pone.0143706] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 11/08/2015] [Indexed: 12/03/2022] Open
Abstract
Background & Objectives It is well known that cognitive impairment in patients with chronic kidney disease (CKD) is characterized by executive dysfunction, rather than memory dysfunction, although the precise mechanism of this remains to be elucidated. The purpose of the present study is to examine the correlation between gray matter volume (GMV) and executive function in CKD patients. Design, Setting, Participants, Measurements This cross-sectional study recruited 95 patients with non-dialysis-dependent CKD (NDD-CKD) with no history of cerebrovascular disease, who underwent brain magnetic resonance imaging (MRI) and Trail Making Test (TMT) in the VCOHP Study. The subjects underwent brain MRI and TMT part A (TMT-A) and part B (TMT-B). The segmentation algorithm from Statistical Parametric Mapping 8 software was applied to every T1-weighted MRI scan to extract tissue maps corresponding to gray matter, white matter, and cerebrospinal fluid. GMV was normalized by dividing by the total intracranial volume, calculated by adding GMV, white matter volume, and cerebrospinal fluid space volume. Then, normalized whole-brain GMV was divided into four categories of brain lobes; frontal, parietal, temporal, and occipital. We assessed the correlation between normalized GMV and TMT using multivariable regression analysis. Results Normalized whole-brain GMV was significantly inversely correlated to the scores of TMT-A, TMT-B, and ΔTMT (TMT-B minus TMT-A). These correlations remained significant even after adjusting for relevant confounding factors. Normalized frontal and temporal GMV, but not parietal and occipital GMV, were significantly inversely correlated with TMT-A, TMT-B, and ΔTMT using multivariable regression analysis. Conclusions The present study demonstrates the correlation between normalized GMV, especially in the frontal and temporal lobes, and executive function, suggesting that fronto-temporal gray matter atrophy might contribute to executive dysfunction in NDD-CKD.
Collapse
Affiliation(s)
- Kazuhiko Tsuruya
- Department of Integrated Therapy for Chronic Kidney Disease, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- * E-mail:
| | - Hisako Yoshida
- Department of Integrated Therapy for Chronic Kidney Disease, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Naoki Haruyama
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kiichiro Fujisaki
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hideki Hirakata
- Division of Nephrology and Dialysis Center, Japanese Red Cross Fukuoka Hospital, Fukuoka, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
148
|
Vieira ER, Mendy A, Prado CM, Gasana J, Albatineh AN. Falls, physical limitations, confusion and memory problems in people with type II diabetes, undiagnosed diabetes and prediabetes, and the influence of vitamins A, D and E. J Diabetes Complications 2015; 29:1159-64. [PMID: 26344725 DOI: 10.1016/j.jdiacomp.2015.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/06/2015] [Accepted: 08/07/2015] [Indexed: 11/16/2022]
Abstract
AIMS To examine the association between type II diabetes, prediabetes and undiagnosed diabetes with falls, physical limitations, confusion and memory problems, and to evaluate the effects of vitamins A, D and E levels on the associations. METHODS Data from 37,973 participants of the National Health and Nutrition Examination Survey was analyzed. RESULTS The participants' mean age was 46±17years, 20% had diabetes of which 17% were unaware of their condition (undiagnosed diabetes), and 21% had prediabetes. Diabetes was significantly associated with falls, difficulties in stooping, crouching, kneeling, completing house chores, getting in and out bed, standing and sitting for long periods, reaching over head, grasping, holding objects, and attending social events. The association between diabetes and confusion or memory problems was stronger for those diagnosed before age 40. Memory problems were reported only by people with diabetes with lower vitamin D levels. Vitamin A and E levels did not modify the association between diabetes and falls or any of the physical functions, confusion or memory problems. Prediabetes was only associated with difficulty standing for long periods. CONCLUSIONS Diabetes was associated with falls, difficulties in physical functioning and attending social events. Vitamin D levels modified the effects on confusion and memory problems.
Collapse
Affiliation(s)
- Edgar R Vieira
- Departments of Physical Therapy & Neuroscience, Florida International University, Miami, Florida, US.
| | - Angelico Mendy
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, Iowa, US
| | - Carla M Prado
- Department of Agricultural, Food & Nutritional Science, & Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Janvier Gasana
- South Florida Asthma Consortium, Fort Lauderdale, Florida, US
| | - Ahmed N Albatineh
- Department of Community Medicine and Behavioral Sciences, Faculty of Medicine, Kuwait University, Kuwait
| |
Collapse
|
149
|
Sato N, Morishita R. The roles of lipid and glucose metabolism in modulation of β-amyloid, tau, and neurodegeneration in the pathogenesis of Alzheimer disease. Front Aging Neurosci 2015; 7:199. [PMID: 26557086 PMCID: PMC4615808 DOI: 10.3389/fnagi.2015.00199] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 10/04/2015] [Indexed: 12/20/2022] Open
Abstract
Diabetes is a risk factor for Alzheimer disease (AD). Apolipoprotein E (ApoE) and several genes related to AD have recently been identified by genome-wide association studies (GWAS) as being closely linked to lipid metabolism. Lipid metabolism and glucose-energy metabolism are closely related. Here, we review the emerging evidence regarding the roles of lipid and glucose metabolism in the modulation of β-amyloid, tau, and neurodegeneration during the pathogenesis of AD. Disruption of homeostasis of lipid and glucose metabolism affects production and clearance of β-amyloid and tau phosphorylation, and induces neurodegeneration. A more integrated understanding of the interactions among lipid, glucose, and protein metabolism is required to elucidate the pathogenesis of AD and to develop next-generation therapeutic options.
Collapse
Affiliation(s)
- Naoyuki Sato
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka UniversitySuita, Japan
- Department of Geriatric Medicine, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka UniversitySuita, Japan
| |
Collapse
|
150
|
Foster-Dingley JC, van der Grond J, Moonen JEF, van den Berg-Huijsmans AA, de Ruijter W, van Buchem MA, de Craen AJM, van der Mast RC. Lower Blood Pressure Is Associated With Smaller Subcortical Brain Volumes in Older Persons. Am J Hypertens 2015; 28:1127-33. [PMID: 25714132 DOI: 10.1093/ajh/hpv006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 01/13/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Both high and low blood pressure (BP) have been positively as well as negatively associated with brain volumes in a variety of populations. The objective of this study was to investigate whether BP is associated with cortical and subcortical brain volumes in older old persons with mild cognitive deficits. METHODS Within the Discontinuation of Antihypertensive Treatment in the Elderly trial, the cross-sectional relation of BP parameters with both cortical and subcortical brain volumes was investigated in 220 older old persons with mild cognitive deficits (43% men, mean age = 80.7 (SD = 4.1), median Mini-Mental State Examination score = 26 (interquartile range: 25-27)), using linear regression analysis. All analyses were adjusted for age, gender, volume of white matter hyperintensities, and duration of antihypertensive treatment. Brain volumes were determined on 3DT1-weighted brain magnetic resonance imaging scans. RESULTS Lower systolic BP, diastolic BP, and mean arterial pressure (MAP) were significantly associated with lower volumes of thalamus and putamen (all P ≤ 0.01). In addition, lower MAP was also associated with reduced hippocampal volume (P = 0.035). There were no associations between any of the BP parameters with cortical gray matter or white matter volume. CONCLUSION In an older population using antihypertensive medication with mild cognitive deficits, a lower BP, rather than a high BP is associated with reduced volumes of thalamus, putamen, and hippocampus.
Collapse
Affiliation(s)
| | - Jeroen van der Grond
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Justine E F Moonen
- Department of Psychiatry, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Wouter de Ruijter
- Department of Public health and Primary care, Leiden University Medical Center, Leiden, the Netherlands
| | - Mark A van Buchem
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Anton J M de Craen
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Roos C van der Mast
- Department of Psychiatry, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|