51
|
Pignalosa FC, Desiderio A, Mirra P, Nigro C, Perruolo G, Ulianich L, Formisano P, Beguinot F, Miele C, Napoli R, Fiory F. Diabetes and Cognitive Impairment: A Role for Glucotoxicity and Dopaminergic Dysfunction. Int J Mol Sci 2021; 22:ijms222212366. [PMID: 34830246 PMCID: PMC8619146 DOI: 10.3390/ijms222212366] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/09/2021] [Accepted: 11/13/2021] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus (DM) is a chronic metabolic disorder characterized by hyperglycemia, responsible for the onset of several long-term complications. Recent evidence suggests that cognitive dysfunction represents an emerging complication of DM, but the underlying molecular mechanisms are still obscure. Dopamine (DA), a neurotransmitter essentially known for its relevance in the regulation of behavior and movement, modulates cognitive function, too. Interestingly, alterations of the dopaminergic system have been observed in DM. This review aims to offer a comprehensive overview of the most relevant experimental results assessing DA’s role in cognitive function, highlighting the presence of dopaminergic dysfunction in DM and supporting a role for glucotoxicity in DM-associated dopaminergic dysfunction and cognitive impairment. Several studies confirm a role for DA in cognition both in animal models and in humans. Similarly, significant alterations of the dopaminergic system have been observed in animal models of experimental diabetes and in diabetic patients, too. Evidence is accumulating that advanced glycation end products (AGEs) and their precursor methylglyoxal (MGO) are associated with cognitive impairment and alterations of the dopaminergic system. Further research is needed to clarify the molecular mechanisms linking DM-associated dopaminergic dysfunction and cognitive impairment and to assess the deleterious impact of glucotoxicity.
Collapse
Affiliation(s)
- Francesca Chiara Pignalosa
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
- URT “Genomic of Diabetes”, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
| | - Antonella Desiderio
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
- URT “Genomic of Diabetes”, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
| | - Paola Mirra
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
- URT “Genomic of Diabetes”, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
| | - Cecilia Nigro
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
- URT “Genomic of Diabetes”, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
| | - Giuseppe Perruolo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
- URT “Genomic of Diabetes”, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
| | - Luca Ulianich
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
- URT “Genomic of Diabetes”, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
| | - Pietro Formisano
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
- URT “Genomic of Diabetes”, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
| | - Francesco Beguinot
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
- URT “Genomic of Diabetes”, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
| | - Claudia Miele
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
- URT “Genomic of Diabetes”, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
- Correspondence: ; Tel.: +39-081-746-3248
| | - Raffaele Napoli
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
| | - Francesca Fiory
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (F.C.P.); (A.D.); (P.M.); (C.N.); (G.P.); (L.U.); (P.F.); (F.B.); (R.N.); (F.F.)
- URT “Genomic of Diabetes”, Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
| |
Collapse
|
52
|
Wu CY, Lin YH, Hsieh HH, Lin JJ, Peng SL. Sex Differences in the Effect of Diabetes on Cerebral Glucose Metabolism. Biomedicines 2021; 9:1661. [PMID: 34829890 PMCID: PMC8615590 DOI: 10.3390/biomedicines9111661] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 12/25/2022] Open
Abstract
The neuroimaging literature indicates that brain structure and function both deteriorate with diabetes, but information on sexual dimorphism in diabetes-related brain alterations is limited. This study aimed to ascertain whether brain metabolism is influenced by sex in an animal model of diabetes. Eleven rats (male, n = 5; female, n = 6) received a single intraperitoneal injection of 70 mg/kg streptozotocin (STZ) to develop diabetes. Another 11 rats (male, n = 5; female, n = 6) received the same amount of solvent through a single intraperitoneal injection. Longitudinal positron emission tomography scans were used to assess cerebral glucose metabolism before and 4 weeks after STZ or solvent administration. Before STZ or solvent injections, there was no evidence of sexual dimorphism in cerebral metabolism (p > 0.05). Compared with healthy control animals, rats with diabetes had significantly decreased brain metabolism in all brain regions (all p < 0.05). In addition, female diabetic rats exhibited further reduction in cerebral metabolism, relative to male diabetic rats (p < 0.05). The results of this study may provide some biological evidence, supporting the existence of a sexual dimorphism in diabetes-related complications.
Collapse
Affiliation(s)
- Chun-Yi Wu
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei Branch, Taipei 112304, Taiwan; (C.-Y.W.); (H.-H.H.)
| | - Yu-Hsin Lin
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei Branch, Taipei 112304, Taiwan;
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404333, Taiwan
| | - Hsin-Hua Hsieh
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei Branch, Taipei 112304, Taiwan; (C.-Y.W.); (H.-H.H.)
| | - Jia-Jia Lin
- Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan 404333, Taiwan;
| | - Shin-Lei Peng
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung 404332, Taiwan
| |
Collapse
|
53
|
Fitzgerald KC, Damian A, Conway D, Mowry EM. Vascular comorbidity is associated with lower brain volumes and lower neuroperformance in a large multiple sclerosis cohort. Mult Scler 2021; 27:1914-1923. [PMID: 33416436 PMCID: PMC8263795 DOI: 10.1177/1352458520984746] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVE The objective of this study is to assess the association between vascular comorbidity burden with clinical and imaging features of disease burden in a large population of people with multiple sclerosis (MS). METHODS We included participants from the MS Partners Advancing Technology Health Solutions (MS PATHS) cohort. We evaluated if vascular comorbidities (diabetes, hypertension, and dyslipidemia) or a composite sum of comorbidities was associated with MS characteristics, including objective neurologic function assessments and quantitative brain magnetic resonance imaging (MRI) measurements in propensity score-weighted models. RESULTS In total, 11,506 participants (6409 (55%) with brain MRI) were included. Individuals with 2+ vascular comorbidities had slower walking speed (standard deviation (SD) = -0.49; 95% confidence interval (CI) = -0.78, -0.19; p = 0.001), slower manual dexterity (SD = -0.41; 95% CI = -0.57, -0.26; p < 0.0001), and fewer correct scores on cognitive processing speed (SD = -0.11; 95% CI = -0.20, -0.02; p = 0.02) versus those with no comorbidities. Those with 2+ had lower brain parenchymal (-0.41%, 95% CI = -0.64, -0.17) and gray matter fractions (-0.30%, 95% CI = -0.49, -0.10), including reduced cortical (-10.10 mL, 95% CI = -15.42, -4.78) and deep (-0.44 mL, 95% CI = -0.84, -0.04) gray matter volumes versus those with no comorbidity. CONCLUSION Increased vascular comorbidity burden was associated with clinical and imaging markers of neurologic dysfunction and neurodegeneration in MS. Strategies to optimize comorbidity management in people with MS are warranted.
Collapse
Affiliation(s)
- Kathryn C Fitzgerald
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA/Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Anne Damian
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Devon Conway
- Mellen Center for Multiple Sclerosis, Cleveland Clinic, Cleveland, OH, USA
| | - Ellen M Mowry
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA/Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
54
|
Dugani SB, Christenson LR, Aakre JA, Bui HH, Vella A, Mielke MM. Association of plasma ceramides with prevalent and incident type 2 diabetes mellitus in middle and older aged adults. Diabetes Res Clin Pract 2021; 179:108991. [PMID: 34333058 PMCID: PMC8478833 DOI: 10.1016/j.diabres.2021.108991] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/15/2021] [Accepted: 07/26/2021] [Indexed: 12/30/2022]
Abstract
AIMS The role of ceramides in the pathogenesis of type 2 diabetes mellitus (T2DM) is incompletely characterized. Given that ceramides represent therapeutic targets to disrupt the euglycemia-T2DM transition, we aimed to characterize their association with prevalent and incident T2DM in a novel cohort. METHODS We examined the cross-sectional and longitudinal association of baseline ceramides with prevalent and incident T2DM among 1423 adults (47% women; median (range) baseline age 72 (51-95) years) in the Mayo Clinic Study of Aging cohort. We examined the associations of ceramides with prevalent T2DM (adjusted odds ratio [95% confidence interval]) at baseline and incident T2DM (adjusted hazard ratio [95% confidence interval]) during median follow-up of 6.2 years, after adjusting for demographic and metabolic factors. RESULTS Among 1423 adults, there were 222 prevalent and 37 incident cases of T2DM. In cross-sectional analyses, higher levels of ceramide C16:0 were associated with lower odds of prevalent T2DM (aOR 0.84 [0.71-0.99];P = 0.03) whereas C18:0 (aOR 1.27 [1.06-1.42];P = 0.01), C18:0/16:0 (aOR 1.41 [1.22-1.62]; P < 0.001) and C18:0/24:0 (aOR 1.22 [1.05-1.41]; P = 0.01) were associated with higher odds. In Cox hazard regression models, C18:0/16:0 (aHR 1.63 [1.26-2.10];P < 0.001) and C18:0 (aHR 1.53 [1.12-2.08];P = 0.01) were associated with increased risk of incident T2DM. CONCLUSIONS In this prospective population-based cohort, ceramides were associated with prevalent T2DM (C16:0,C18:0, C18:0/C16:0 ratio, C18:0/C24:0 ratio) and incident T2DM (C18:0, C18:0/C16:0 ratio) and could suggest targets for the primary and secondary prevention of T2DM.
Collapse
Affiliation(s)
- Sagar B Dugani
- Division of Hospital Internal Medicine, Mayo Clinic, Rochester, MN, United States; Division of Health Care Delivery Research, Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN, United States
| | - Luke R Christenson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Jeremiah A Aakre
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Hai H Bui
- Eli Lilly and Company, Indianapolis, IN, United States
| | - Adrian Vella
- Division of Endocrinology, Mayo Clinic, Rochester, MN, United States
| | - Michelle M Mielke
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States; Department of Neurology, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
55
|
Chen Y, Wang X, Guan L, Wang Y. Role of White Matter Hyperintensities and Related Risk Factors in Vascular Cognitive Impairment: A Review. Biomolecules 2021; 11:biom11081102. [PMID: 34439769 PMCID: PMC8391787 DOI: 10.3390/biom11081102] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/24/2021] [Accepted: 07/25/2021] [Indexed: 02/06/2023] Open
Abstract
White matter hyperintensities (WMHs) of presumed vascular origin are one of the imaging markers of cerebral small-vessel disease, which is prevalent in older individuals and closely associated with the occurrence and development of cognitive impairment. The heterogeneous nature of the imaging manifestations of WMHs creates difficulties for early detection and diagnosis of vascular cognitive impairment (VCI) associated with WMHs. Because the underlying pathological processes and biomarkers of WMHs and their development in cognitive impairment remain uncertain, progress in prevention and treatment is lagging. For this reason, this paper reviews the status of research on the features of WMHs related to VCI, as well as mediators associated with both WMHs and VCI, and summarizes potential treatment strategies for the prevention and intervention in WMHs associated with VCI.
Collapse
Affiliation(s)
- Yiyi Chen
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (Y.C.); (X.W.)
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing 100070, China
| | - Xing Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (Y.C.); (X.W.)
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing 100070, China
- Department of Neurology, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing 400000, China
| | - Ling Guan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (Y.C.); (X.W.)
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing 100070, China
- Correspondence: (L.G.); (Y.W.)
| | - Yilong Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (Y.C.); (X.W.)
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing 100070, China
- Correspondence: (L.G.); (Y.W.)
| |
Collapse
|
56
|
Al-Anbari HSN, Ismail DK, Hasan MK, Aga QAAK, Shinu P, Nair AB. High Blood Lead Levels: An Increased Risk for Development of Brain Hyperintensities among Type 2 Diabetes Mellitus Patients. Biol Trace Elem Res 2021; 199:2149-2157. [PMID: 32865724 DOI: 10.1007/s12011-020-02359-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/25/2020] [Indexed: 01/21/2023]
Abstract
The current study was aimed to ascertain the effect of blood lead level on brain tissues in patients with type 2 diabetes. A total of 300 human participants ages 27 to 60 years with type 2 diabetes (n = 150) and healthy individuals (n = 150) were included in this study. The serum samples were used for measuring HbA1c and fasting blood glucose. Blood lead level was measured using flame atomic absorption spectrophotometer. Magnetic resonance imaging sub-analysis was used to assess the brain hyperintensities. Brain hyperintensities were found in 55% of patients with diabetes and 6% of non-diabetic control group subjects. The deep white matter hyperintensities were observed in 45% of diabetic patients, while the subcortical hyperintensities were noted in 10% of cases. Entorhinal cortex changes (31%) and hippocampus changes (42%) were noted in diabetic patients with brain hyperintensities. Diabetic patients with brain hyperintensities showed higher blood lead levels, HbA1c, and fasting blood sugar (p < 0.0001) as compared with healthy volunteers. A higher correlation (R2 = 0.8922) was found between deep white matter hyperintensities' size and blood lead levels. In nutshell, persistence of high blood lead level in diabetic patients may progress to brain hyperintensities which may consequently lead to cognitive, behavioral changes and Alzheimer's disease.
Collapse
Affiliation(s)
| | - Dawser K Ismail
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Al Esraa University College, Baghdad, 10069, Iraq
| | - Mohammed Khudair Hasan
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Al Esraa University College, Baghdad, 10069, Iraq
| | - Qutaiba Ahmed Al Khames Aga
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Philadelphia University, P.O. Box 1, Amman, 19392, Jordan.
| | - Pottathil Shinu
- Department of Biomedical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, 31982, Saudi Arabia
| | - Anroop B Nair
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, 31982, Saudi Arabia
| |
Collapse
|
57
|
Yaffe K, Vittinghoff E, Hoang T, Matthews K, Golden SH, Zeki Al Hazzouri A. Cardiovascular Risk Factors Across the Life Course and Cognitive Decline: A Pooled Cohort Study. Neurology 2021; 96:e2212-e2219. [PMID: 33731482 PMCID: PMC8166431 DOI: 10.1212/wnl.0000000000011747] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 01/28/2021] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Cardiovascular risk factors (CVRFs) are associated with increased risk of cognitive decline, but little is known about how early adult CVRFs and those across the life course might influence late-life cognition. To test the hypothesis that CVRFs across the adult life course are associated with late-life cognitive changes, we pooled data from 4 prospective cohorts (n = 15,001, ages 18-95). METHODS We imputed trajectories of body mass index (BMI), fasting glucose (FG), systolic blood pressure (SBP), and total cholesterol (TC) for older adults. We used linear mixed models to determine the association of early adult, midlife, and late-life CVRFs with late-life decline on global cognition (Modified Mini-Mental State Examination [3MS]) and processing speed (Digit Symbol Substitution Test [DSST]), adjusting for demographics, education, and cohort. RESULTS Elevated BMI, FG, and SBP (but not TC) at each time period were associated with greater late-life decline. Early life CVRFs were associated with the greatest change, an approximate doubling of mean 10-year decline (an additional 3-4 points for 3MS or DSST). Late-life CVRFs were associated with declines in early late life (<80 years) but with gains in very late life (≥80 years). After adjusting for CVRF exposures at all time periods, the associations for early adult and late-life CVRFs persisted. CONCLUSIONS We found that imputed CVRFs across the life course, especially in early adulthood, were associated with greater late-life cognitive decline. Our results suggest that CVRF treatment in early adulthood could benefit late-life cognition, but that treatment in very late life may not be as helpful for these outcomes.
Collapse
Affiliation(s)
- Kristine Yaffe
- From the Departments of Psychiatry and Neurology (K.Y.) and Epidemiology & Biostatistics (K.Y., E.V.), University of California San Francisco; San Francisco Veterans Affairs Medical Center (K.Y.); Northern California Institute Research for Research and Education (T.H.), San Francisco; Department of Psychiatry (K.M.), University of Pittsburgh, PA; Department of Medicine (S.H.G.), Johns Hopkins University School of Medicine; Department of Epidemiology (S.H.G.), Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD; and Department of Epidemiology (A.Z.A.H.), Mailman School of Public Health, Columbia University, New York, NY.
| | - Eric Vittinghoff
- From the Departments of Psychiatry and Neurology (K.Y.) and Epidemiology & Biostatistics (K.Y., E.V.), University of California San Francisco; San Francisco Veterans Affairs Medical Center (K.Y.); Northern California Institute Research for Research and Education (T.H.), San Francisco; Department of Psychiatry (K.M.), University of Pittsburgh, PA; Department of Medicine (S.H.G.), Johns Hopkins University School of Medicine; Department of Epidemiology (S.H.G.), Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD; and Department of Epidemiology (A.Z.A.H.), Mailman School of Public Health, Columbia University, New York, NY
| | - Tina Hoang
- From the Departments of Psychiatry and Neurology (K.Y.) and Epidemiology & Biostatistics (K.Y., E.V.), University of California San Francisco; San Francisco Veterans Affairs Medical Center (K.Y.); Northern California Institute Research for Research and Education (T.H.), San Francisco; Department of Psychiatry (K.M.), University of Pittsburgh, PA; Department of Medicine (S.H.G.), Johns Hopkins University School of Medicine; Department of Epidemiology (S.H.G.), Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD; and Department of Epidemiology (A.Z.A.H.), Mailman School of Public Health, Columbia University, New York, NY
| | - Karen Matthews
- From the Departments of Psychiatry and Neurology (K.Y.) and Epidemiology & Biostatistics (K.Y., E.V.), University of California San Francisco; San Francisco Veterans Affairs Medical Center (K.Y.); Northern California Institute Research for Research and Education (T.H.), San Francisco; Department of Psychiatry (K.M.), University of Pittsburgh, PA; Department of Medicine (S.H.G.), Johns Hopkins University School of Medicine; Department of Epidemiology (S.H.G.), Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD; and Department of Epidemiology (A.Z.A.H.), Mailman School of Public Health, Columbia University, New York, NY
| | - Sherita H Golden
- From the Departments of Psychiatry and Neurology (K.Y.) and Epidemiology & Biostatistics (K.Y., E.V.), University of California San Francisco; San Francisco Veterans Affairs Medical Center (K.Y.); Northern California Institute Research for Research and Education (T.H.), San Francisco; Department of Psychiatry (K.M.), University of Pittsburgh, PA; Department of Medicine (S.H.G.), Johns Hopkins University School of Medicine; Department of Epidemiology (S.H.G.), Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD; and Department of Epidemiology (A.Z.A.H.), Mailman School of Public Health, Columbia University, New York, NY
| | - Adina Zeki Al Hazzouri
- From the Departments of Psychiatry and Neurology (K.Y.) and Epidemiology & Biostatistics (K.Y., E.V.), University of California San Francisco; San Francisco Veterans Affairs Medical Center (K.Y.); Northern California Institute Research for Research and Education (T.H.), San Francisco; Department of Psychiatry (K.M.), University of Pittsburgh, PA; Department of Medicine (S.H.G.), Johns Hopkins University School of Medicine; Department of Epidemiology (S.H.G.), Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD; and Department of Epidemiology (A.Z.A.H.), Mailman School of Public Health, Columbia University, New York, NY
| |
Collapse
|
58
|
Melo HM, Seixas da Silva GDS, Sant'Ana MR, Teixeira CVL, Clarke JR, Miya Coreixas VS, de Melo BC, Fortuna JTS, Forny-Germano L, Ledo JH, Oliveira MS, Figueiredo CP, Pardossi-Piquard R, Checler F, Delgado-García JM, Gruart A, Velloso LA, Balthazar MLF, Cintra DE, Ferreira ST, De Felice FG. Palmitate Is Increased in the Cerebrospinal Fluid of Humans with Obesity and Induces Memory Impairment in Mice via Pro-inflammatory TNF-α. Cell Rep 2021; 30:2180-2194.e8. [PMID: 32075735 DOI: 10.1016/j.celrep.2020.01.072] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 10/08/2019] [Accepted: 01/22/2020] [Indexed: 12/26/2022] Open
Abstract
Obesity has been associated with cognitive decline, atrophy of brain regions related to learning and memory, and higher risk of developing dementia. However, the molecular mechanisms underlying these neurological alterations are still largely unknown. Here, we investigate the effects of palmitate, a saturated fatty acid present at high amounts in fat-rich diets, in the brain. Palmitate is increased in the cerebrospinal fluid (CSF) of overweight and obese patients with amnestic mild cognitive impairment. In mice, intracerebroventricular infusion of palmitate impairs synaptic plasticity and memory. Palmitate induces astroglial and microglial activation in the mouse hippocampus, and its deleterious impact is mediated by microglia-derived tumor necrosis factor alpha (TNF-α) signaling. Our results establish that obesity is associated with increases in CSF palmitate. By defining a pro-inflammatory mechanism by which abnormal levels of palmitate in the brain impair memory, the results further suggest that anti-inflammatory strategies may attenuate memory impairment in obesity.
Collapse
Affiliation(s)
- Helen M Melo
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Gisele da S Seixas da Silva
- Federal Institute of Education, Science and Technology of Rio de Janeiro, Rio de Janeiro, RJ 20270-021, Brazil
| | - Marcella Ramos Sant'Ana
- Laboratory of Nutritional Genomics (LabGeN), School of Applied Sciences and CELN - Nutrigenomics and Lipids Research Center, School of Applied Sciences, University of Campinas (UNICAMP), Limeira, SP 13484-350, Brazil
| | - Camila Vieira Ligo Teixeira
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN) and Department of Neurology, Neuroimaging Laboratory, University of Campinas (UNICAMP), Campinas, SP 13083-887, Brazil
| | - Julia R Clarke
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Vivian S Miya Coreixas
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Bruno C de Melo
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Juliana T S Fortuna
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Leticia Forny-Germano
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - José Henrique Ledo
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Maíra S Oliveira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Claudia P Figueiredo
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Raphaelle Pardossi-Piquard
- Université Côte d'Azur, INSERM, CNRS/UMR7275, IPMC, team labeled "Laboratory of Excellence (LABEX) Distalz," 660 route des Lucioles, 06560 Sophia-Antipolis, Valbonne, France
| | - Frédéric Checler
- Université Côte d'Azur, INSERM, CNRS/UMR7275, IPMC, team labeled "Laboratory of Excellence (LABEX) Distalz," 660 route des Lucioles, 06560 Sophia-Antipolis, Valbonne, France
| | | | - Agnès Gruart
- Division of Neuroscience, Pablo de Olavide University, Seville 41013, Spain
| | - Licio A Velloso
- Laboratory of Cell Signalling, Obesity and Comorbidities Research Centre, University of Campinas, Campinas, SP 13084-761, Brazil
| | - Marcio L F Balthazar
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN) and Department of Neurology, Neuroimaging Laboratory, University of Campinas (UNICAMP), Campinas, SP 13083-887, Brazil
| | - Dennys E Cintra
- Laboratory of Nutritional Genomics (LabGeN), School of Applied Sciences and CELN - Nutrigenomics and Lipids Research Center, School of Applied Sciences, University of Campinas (UNICAMP), Limeira, SP 13484-350, Brazil
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil; Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil; Centre for Neuroscience Studies and Department of Psychiatry, Queen's University, Kingston, ON K7L 3N6, Canada.
| |
Collapse
|
59
|
Shinohara M, Kikuchi M, Onishi-Takeya M, Tashiro Y, Suzuki K, Noda Y, Takeda S, Mukouzono M, Nagano S, Fukumori A, Morishita R, Nakaya A, Sato N. Upregulated expression of a subset of genes in APP; ob/ ob mice: Evidence of an interaction between diabetes-linked obesity and Alzheimer's disease. FASEB Bioadv 2021; 3:323-333. [PMID: 33977233 PMCID: PMC8103720 DOI: 10.1096/fba.2020-00151] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/17/2021] [Accepted: 01/29/2021] [Indexed: 12/26/2022] Open
Abstract
Clinical studies have indicated that obesity and diabetes are associated with Alzheimer's disease (AD) and neurodegeneration. However, the mechanism by which obesity/diabetes and AD interact with each other and contribute to dementia remains elusive. To obtain insights into their interaction at molecular levels, we performed gene expression analysis of APP;ob/ob mice, which were generated by crossing transgenic AD model mice (APP23 mice) with ob/ob mice, which are obese and mildly diabetic. The Aβ level in these mice was reduced compared with that in pure APP mice. However, we identified a cluster of genes (cluster 10) upregulated in APP;ob/ob mice but not in either APP or ob/ob mice. Interestingly, genes upregulated in the human AD brain were enriched in cluster 10. Moreover, genes in cluster 10 formed a network and shared upregulated genes with a cell model of neurodegeneration and other models of neurological disorders such as ischemia and epilepsy. In silico analyses showed that serum response factor (SRF), recently identified in a single-cell analysis of human brains as a transcription factor that can control the conversion from healthy cells to AD cells, might be a common transcriptional regulator for a subset of cluster 10 genes. These data suggest that upregulation of genes uniquely associated with APP;ob/ob mice is an evidence of the interaction between obesity/diabetes and AD.
Collapse
Affiliation(s)
- Mitsuru Shinohara
- Department of Aging Neurobiology Center for Development of Advanced Medicine for Dementia National Center for Geriatrics and Gerontology Obu Aichi Japan.,Department of Aging Neurobiology Graduate School of Medicine Osaka University Osaka Japan
| | - Masataka Kikuchi
- Department of Genome Informatics Graduate School of Medicine Osaka University Osaka Japan
| | - Miyuki Onishi-Takeya
- Department of Geriatric Medicine Graduate School of Medicine Osaka University Osaka Japan
| | - Yoshitaka Tashiro
- Department of Aging Neurobiology Center for Development of Advanced Medicine for Dementia National Center for Geriatrics and Gerontology Obu Aichi Japan
| | - Kaoru Suzuki
- Department of Aging Neurobiology Center for Development of Advanced Medicine for Dementia National Center for Geriatrics and Gerontology Obu Aichi Japan
| | - Yasuhiro Noda
- Department of Aging Neurobiology Center for Development of Advanced Medicine for Dementia National Center for Geriatrics and Gerontology Obu Aichi Japan
| | - Shuko Takeda
- Department of Clinical Gene Therapy Graduate School of Medicine Osaka University Osaka Japan
| | - Masahiro Mukouzono
- Department of Clinical Gene Therapy Graduate School of Medicine Osaka University Osaka Japan
| | - Seiichi Nagano
- Department of Neurology Graduate School of Medicine Osaka University Osaka Japan
| | - Akio Fukumori
- Department of Aging Neurobiology Center for Development of Advanced Medicine for Dementia National Center for Geriatrics and Gerontology Obu Aichi Japan.,Department of Aging Neurobiology Graduate School of Medicine Osaka University Osaka Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy Graduate School of Medicine Osaka University Osaka Japan
| | - Akihiro Nakaya
- Department of Genome Informatics Graduate School of Medicine Osaka University Osaka Japan
| | - Naoyuki Sato
- Department of Aging Neurobiology Center for Development of Advanced Medicine for Dementia National Center for Geriatrics and Gerontology Obu Aichi Japan.,Department of Aging Neurobiology Graduate School of Medicine Osaka University Osaka Japan
| |
Collapse
|
60
|
Gerstein HC, Smith EE, Ramasundarahettige C, Desai D, Awadalla P, Broet P, Black S, Dummer TJB, Hicks J, Moody A, Tardif JC, Teo KK, Vena J, Yusuf S, Lee DS, Friedrich MG, Anand SS. Diabetes, Brain Infarcts, Cognition, and Small Vessels in the Canadian Alliance for Healthy Hearts and Minds Study. J Clin Endocrinol Metab 2021; 106:e891-e898. [PMID: 33165530 PMCID: PMC7823245 DOI: 10.1210/clinem/dgaa815] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Diabetes is a risk factor for cerebrovascular disease and cognitive impairment. The anatomical basis for this is uncertain. METHODS The Canadian Alliance for Healthy Hearts and Minds collected brain and carotid magnetic resonance imaging (MRI) and 2 cognitive tests (the Digit Symbol Substitution Test and the Montreal Cognitive Assessment test) in a cross-sectional sample of men and women. Brain MRIs identified brain infarcts (BI), lacunar BI, high white matter hyperintensity (WMH), vascular brain injury (VBI; BI or high WMH), and small vessel VBI (lacunar BI or high WMH). Carotid MRIs estimated carotid wall volume, a measure of subclinical atherosclerosis. Cognitive scores were standardized to each site's mean score, and cognitive impairment was identified by 1 or both test scores ≤1 standard deviation below the site's mean score on that test. RESULTS The 7733 participants included 495 participants (6.4%) with diabetes, of whom 388 were taking diabetes drugs. After age and sex adjustment, diabetes was independently associated with BI (odds ratio [OR] 1.53, 95% confidence interval [CI] 1.05, 2.24), VBI (OR 1.64, 95% CI 1.26, 2.13), small vessel VBI (OR 1.67, 95% CI 1.28, 2.19), and cognitive impairment (OR 1.47, 95% CI 1.20, 1.80). The association between diabetes and small vessel VBI persisted after adjustment for cerebrovascular disease risk factors and nonlacunar infarcts (OR 1.52, 95% CI 1.15, 2.01), and the association with cognitive impairment persisted after adjustment for small vessel VBI (OR 1.27, 95% CI 1.03, 1.56). CONCLUSION Small vessel disease characterizes much of the relationship between diabetes and VBI. However, additional factors are required to disentangle the relationship between diabetes and cognitive impairment.
Collapse
Affiliation(s)
- Hertzel C Gerstein
- Population Health Research Institute, Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Department of Health Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- Correspondence and Reprint Requests: Hertzel C. Gerstein, Department of Medicine, HSC 3V38, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada. E-mail:
| | - Eric E Smith
- Hotchkiss Brain Institute, Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Chinthanie Ramasundarahettige
- Population Health Research Institute, Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Dipika Desai
- Population Health Research Institute, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Philip Awadalla
- Department of Molecular Genetics, Ontario Institute for Cancer Research, University of Toronto, Toronto, Ontario, Canada
| | - Philippe Broet
- Department of Preventive and Social Medicine, École de santé publique, Université de Montréal, Montreal, Quebec, Canada
- Research Centre, CHU Sainte Justine, Montreal, Quebec, Canada
| | - Sandra Black
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Trevor J B Dummer
- Cancer Control Research, BC Cancer, and the School of Population and Public Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jason Hicks
- Atlantic PATH, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Alan Moody
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Jean-Claude Tardif
- Department of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| | - Koon K Teo
- Population Health Research Institute, Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Department of Health Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Jennifer Vena
- Alberta’s Tomorrow Project, Cancer Research and Analytics, Cancer Control Alberta, Alberta Health Services, Edmonton, Alberta, Canada
| | - Salim Yusuf
- Population Health Research Institute, Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Department of Health Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Douglas S Lee
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
- Peter Munk Cardiac Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Matthias G Friedrich
- Department of Medicine and Diagnostic Radiology, McGill University, Montreal, Quebec, Canada
| | - Sonia S Anand
- Population Health Research Institute, Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Department of Health Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
61
|
Abstract
The American Diabetes Association (ADA) "Standards of Medical Care in Diabetes" includes the 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 (https://doi.org/10.2337/dc21-SPPC), 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 (https://doi.org/10.2337/dc21-SINT). Readers who wish to comment on the Standards of Care are invited to do so at professional.diabetes.org/SOC.
Collapse
|
62
|
Zlokovic BV, Gottesman RF, Bernstein KE, Seshadri S, McKee A, Snyder H, Greenberg SM, Yaffe K, Schaffer CB, Yuan C, Hughes TM, Daemen MJ, Williamson JD, González HM, Schneider J, Wellington CL, Katusic ZS, Stoeckel L, Koenig JI, Corriveau RA, Fine L, Galis ZS, Reis J, Wright JD, Chen J. Vascular contributions to cognitive impairment and dementia (VCID): A report from the 2018 National Heart, Lung, and Blood Institute and National Institute of Neurological Disorders and Stroke Workshop. Alzheimers Dement 2020; 16:1714-1733. [PMID: 33030307 DOI: 10.1002/alz.12157] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/30/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022]
Abstract
Vascular contributions to cognitive impairment and dementia (VCID) are characterized by the aging neurovascular unit being confronted with and failing to cope with biological insults due to systemic and cerebral vascular disease, proteinopathy including Alzheimer's biology, metabolic disease, or immune response, resulting in cognitive decline. This report summarizes the discussion and recommendations from a working group convened by the National Heart, Lung, and Blood Institute and the National Institute of Neurological Disorders and Stroke to evaluate the state of the field in VCID research, identify research priorities, and foster collaborations. As discussed in this report, advances in understanding the biological mechanisms of VCID across the wide spectrum of pathologies, chronic systemic comorbidities, and other risk factors may lead to potential prevention and new treatment strategies to decrease the burden of dementia. Better understanding of the social determinants of health that affect risks for both vascular disease and VCID could provide insight into strategies to reduce racial and ethnic disparities in VCID.
Collapse
Affiliation(s)
| | | | | | - Sudha Seshadri
- University of Texas Health Science Center, San Antonio and Boston University, San Antonio, Texas, USA
| | - Ann McKee
- VA Boston Healthcare System and Boston University, Boston, Massachusetts, USA
| | | | - Steven M Greenberg
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kristine Yaffe
- University of California, San Francisco, San Francisco, California, USA
| | | | - Chun Yuan
- University of Washington, Seattle, Washington, USA
| | - Timothy M Hughes
- Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Mat J Daemen
- Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | | | | | | | | | | | - Luke Stoeckel
- National Institute on Aging, Bethesda, Maryland, USA
| | - James I Koenig
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA
| | - Roderick A Corriveau
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, USA
| | - Lawrence Fine
- National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Zorina S Galis
- National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | - Jared Reis
- National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| | | | - Jue Chen
- National Heart, Lung, and Blood Institute, Bethesda, Maryland, USA
| |
Collapse
|
63
|
Common Brain Structural Alterations Associated with Cardiovascular Disease Risk Factors and Alzheimer's Dementia: Future Directions and Implications. Neuropsychol Rev 2020; 30:546-557. [PMID: 33011894 DOI: 10.1007/s11065-020-09460-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 09/24/2020] [Indexed: 01/18/2023]
Abstract
Recent reports suggest declines in the age-specific risk of Alzheimer's dementia in higher income Western countries. At the same time, investigators believe that worldwide trends of increasing mid-life modifiable risk factors [e.g., cardiovascular disease (CVD) risk factors] coupled with the growth of the world's oldest age groups may nonetheless lead to an increase in Alzheimer's dementia. Thus, understanding the overlap in neuroanatomical profiles associated with CVD risk factors and AD may offer more relevant targets for investigating ways to reduce the growing dementia epidemic than current targets specific to isolated AD-related neuropathology. We hypothesized that a core group of common brain structural alterations exist between CVD risk factors and Alzheimer's dementia. Two co-authors conducted independent literature reviews in PubMed using search terms for CVD risk factor burden (separate searches for 'cardiovascular disease risk factors', 'hypertension', and 'Type 2 diabetes') and 'aging' or 'Alzheimer's dementia' with either 'grey matter volumes' or 'white matter'. Of studies that reported regionally localized results, we found support for our hypothesis, determining 23 regions commonly associated with both CVD risk factors and Alzheimer's dementia. Within this context, we outline future directions for research as well as larger cerebrovascular implications for these commonalities. Overall, this review supports previous as well as more recent calls for the consideration that both vascular and neurodegenerative factors contribute to the pathogenesis of dementia.
Collapse
|
64
|
Human APRIL and FGF-21 and adhesion molecules in relation to cognitive function in elderly diabetic patients. Int J Diabetes Dev Ctries 2020. [DOI: 10.1007/s13410-020-00832-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
65
|
Ha BJ, Bae IS, Kim JM, Cheong JH, Ryu JI, Han MH. Effects of Possible Osteoporotic Conditions on the Recurrence of Chronic Subdural Hematoma. Front Neurol 2020; 11:538257. [PMID: 33071940 PMCID: PMC7542308 DOI: 10.3389/fneur.2020.538257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 08/27/2020] [Indexed: 01/30/2023] Open
Abstract
The recurrence rate of chronic subdural hematoma (CSDH) has been reported to range from 2.3 to 33%. As bridging veins are composed of abundant collagen bundles and bone matrix, we aimed to investigate the possible associations between skull Hounsfield unit (HU) values and the recurrence of CSDH. We retrospectively enrolled patients with CSDH who underwent burr hole surgery. The HU values of the frontal skull were measured on brain CT scans. The cumulative hazard for recurrence was estimated according to predictive factors. To identify the independent predictors associated with the recurrence of CSDH, hazard ratios (HRs) were estimated using multivariate Cox regression analysis. A total of 208 consecutive patients who underwent burr hole trephination for CSDH over a 7-years period at a single institution were enrolled in this study. We found that age, greater midline shift (≥10.5 mm), lower skull HU (<769.5), and diabetes were independent predictors for the recurrence of CSDH (HR 1.06, 95% confidence interval [CI] 1.00–1.12, p = 0.042; HR 5.37, 95% CI 1.48–19.46, p = 0.010; HR 6.71, 95% CI 1.84–24.45, p = 0.004; and HR 3.30, 95% CI 1.05–10.43, p = 0.042, respectively). A relationship between possible low bone mineral density (BMD) and CSDH recurrence was observed. In addition, age, greater preoperative midline shift, and diabetes were also identified as predictive factors for recurrence. We expect that our findings may facilitate our understanding of the possible association between CSDH and BMD.
Collapse
Affiliation(s)
- Byeong Jin Ha
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri, South Korea
| | - In-Suk Bae
- Department of Neurosurgery, Eulji University Hospital, Seoul, South Korea
| | - Jae Min Kim
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri, South Korea
| | - Jin Hwan Cheong
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri, South Korea
| | - Je Il Ryu
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri, South Korea
| | - Myung-Hoon Han
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri, South Korea
| |
Collapse
|
66
|
Hay M, Barnes C, Huentelman M, Brinton R, Ryan L. Hypertension and Age-Related Cognitive Impairment: Common Risk Factors and a Role for Precision Aging. Curr Hypertens Rep 2020; 22:80. [PMID: 32880739 PMCID: PMC7467861 DOI: 10.1007/s11906-020-01090-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Purpose of Review Precision Aging® is a novel concept that we have recently employed to describe how the model of precision medicine can be used to understand and define the multivariate risks that drive age-related cognitive impairment (ARCI). Hypertension and cardiovascular disease are key risk factors for both brain function and cognitive aging. In this review, we will discuss the common mechanisms underlying the risk factors for both hypertension and ARCI and how the convergence of these mechanisms may be amplified in an individual to drive changes in brain health and accelerate cognitive decline. Recent Findings Currently, our cognitive health span does not match our life span. Age-related cognitive impairment and preventing and treating ARCI will require an in-depth understanding of the interrelated risk factors, including individual genetic profiles, that affect brain health and brain aging. Hypertension and cardiovascular disease are important risk factors for ARCI. And, many of the risk factors for developing hypertension, such as diabetes, smoking, stress, viral infection, and age, are shared with the development of ARCI. We must first understand the mechanisms common to the converging risk factors in hypertension and ARCI and then design person-specific therapies to optimize individual brain health. Summary The understanding of the convergence of shared risk factors between hypertension and ARCI is required to develop individualized interventions to optimize brain health across the life span. We will conclude with a discussion of possible steps that may be taken to decrease ARCI and optimize an individual’s cognitive life span.
Collapse
Affiliation(s)
- Meredith Hay
- Department of Physiology, University of Arizona, 1501 N Campbell Rd, Room 4103, Tucson, AZ, 85724, USA.
- Psychology Department, University of Arizona, Tucson, AZ, USA.
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA.
| | - Carol Barnes
- Psychology Department, University of Arizona, Tucson, AZ, USA
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA
| | - Matt Huentelman
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA
- Neurogenomics Division, TGen, Phoenix, AZ, USA
| | - Roberta Brinton
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA
- Center for Innovative Brain Sciences, University of Arizona, Tucson, AZ, USA
| | - Lee Ryan
- Psychology Department, University of Arizona, Tucson, AZ, USA
- Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
67
|
Jaleel MA, Ramesh S, Raju S, Sharma R, Anjankar S, Reddy RH. Cognitive Impairment in Moderate Degree Diffuse Axonal Head Injuries: Analysis of 84 Cases Using MMSE. INDIAN JOURNAL OF NEUROTRAUMA 2020. [DOI: 10.1055/s-0039-1700365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Abstract
Background Cognitive impairment is commonly seen in traumatic brain injury survivors. Posttraumatic cognitive sequelae may be more devastating than focal motor and focal sensory deficits, and are usually left unattended.
Aim and Objective Aim of this study was to assess cognitive impairment in patients who had sustained moderate degree diffuse axonal injuries and having good outcome (Glasgow Outcome Score of 5).
Methods and Materials Prospective observational study was done from 2011 to 2015 on the patients who had sustained moderated degree diffuse brain injuries. Eighty-four cases fulfilling the inclusion criteria were studied. Patients were assessed with Mini-Mental Status Examination at discharge, end of 1 month, and at 3 months.
Result Seventy-six were males and 8 were females. Age ranged from 16 to 49 years. Note that 4.76% (4) patients had hypotension at presentation, 32.14% (27) patients had associated injuries, and 19.04% (16) patients had hyponatremia at presentation. Diabetes mellitus was seen in 34.52% (29) patients, while hypertension was seen in 14.28% (12). At 3 months’ follow-up, 19.06% (16) patients had cognitive impairment. The present study revealed that hypotension and presence of associated injuries at presentation raises the odds of having cognitive impairment by 8 and 5 times, respectively.
Conclusion Routine assessment of cognitive impairment in head injury survivors is essential as it may help in identifying cognitive deficits. Early intervention of neurorehabilitation to such patients results in better neurocognitive outcome. Hypotension and associated injuries are associated with poor cognitive outcome.
Collapse
Affiliation(s)
- Momin Abdul Jaleel
- Department of Neurosurgery, Kamineni Hospitals, Hyderabad, Telangana, India
| | - Shighakolli Ramesh
- Department of Neurosurgery, Kamineni Hospitals, Hyderabad, Telangana, India
| | - Subodh Raju
- Department of Neurosurgery, Apollo Hospitals, Hyderabad, Telangana, India
| | - Renuka Sharma
- Department of Neurosurgery, Kamineni Hospitals, Hyderabad, Telangana, India
| | | | | |
Collapse
|
68
|
Brodtmann A, Khlif MS, Egorova N, Veldsman M, Bird LJ, Werden E. Dynamic Regional Brain Atrophy Rates in the First Year After Ischemic Stroke. Stroke 2020; 51:e183-e192. [PMID: 32772680 DOI: 10.1161/strokeaha.120.030256] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND PURPOSE Brain atrophy can be regarded as an end-organ effect of cumulative cardiovascular risk factors. Accelerated brain atrophy is described following ischemic stroke, but it is not known whether atrophy rates vary over the poststroke period. Examining rates of brain atrophy allows the identification of potential therapeutic windows for interventions to prevent poststroke brain atrophy. METHODS We charted total and regional brain volume and cortical thickness trajectories, comparing atrophy rates over 2 time periods in the first year after ischemic stroke: within 3 months (early period) and between 3 and 12 months (later period). Patients with first-ever or recurrent ischemic stroke were recruited from 3 Melbourne hospitals at 1 of 2 poststroke time points: within 6 weeks (baseline) or 3 months. Whole-brain 3T magnetic resonance imaging was performed at 3 time points: baseline, 3 months, and 12 months. Eighty-six stroke participants completed testing at baseline; 125 at 3 months (76 baseline follow-up plus 49 delayed recruitment); and 113 participants at 12 months. Their data were compared with 40 healthy control participants with identical testing. We examined 5 brain measures: hippocampal volume, thalamic volume, total brain and hemispheric brain volume, and cortical thickness. We tested whether brain atrophy rates differed between time points and groups. A linear mixed-effect model was used to compare brain structural changes, including age, sex, years of education, a composite cerebrovascular risk factor score, and total intracranial volume as covariates. RESULTS Atrophy rates were greater in stroke than control participants. Ipsilesional hemispheric, hippocampal, and thalamic atrophy rates were 2 to 4 times greater in the early versus later period. CONCLUSIONS Regional atrophy rates vary over the first year after stroke. Rapid brain volume loss in the first 3 months after stroke may represent a potential window for intervention. Registration: URL: https://www.clinicaltrials.gov. Unique identifier: NCT02205424.
Collapse
Affiliation(s)
- Amy Brodtmann
- The Florey Institute of Neuroscience and Mental Health (A.B., M.S.K., N.E., M.V., L.J.B., E.W.), University of Melbourne, Australia.,Melbourne Dementia Research Centre, Florey Institute (A.B., N.E., E.W.), University of Melbourne, Australia.,Eastern Cognitive Disorders Clinic, Eastern Health, Monash University, Australia (A.B.).,Department of Neurology, Austin Health, Melbourne, Australia (A.B.)
| | - Mohamed Salah Khlif
- The Florey Institute of Neuroscience and Mental Health (A.B., M.S.K., N.E., M.V., L.J.B., E.W.), University of Melbourne, Australia
| | - Natalia Egorova
- The Florey Institute of Neuroscience and Mental Health (A.B., M.S.K., N.E., M.V., L.J.B., E.W.), University of Melbourne, Australia.,Melbourne Dementia Research Centre, Florey Institute (A.B., N.E., E.W.), University of Melbourne, Australia.,Melbourne School of Psychological Sciences (N.E.), University of Melbourne, Australia
| | - Michele Veldsman
- The Florey Institute of Neuroscience and Mental Health (A.B., M.S.K., N.E., M.V., L.J.B., E.W.), University of Melbourne, Australia
| | - Laura J Bird
- The Florey Institute of Neuroscience and Mental Health (A.B., M.S.K., N.E., M.V., L.J.B., E.W.), University of Melbourne, Australia
| | - Emilio Werden
- The Florey Institute of Neuroscience and Mental Health (A.B., M.S.K., N.E., M.V., L.J.B., E.W.), University of Melbourne, Australia.,Melbourne Dementia Research Centre, Florey Institute (A.B., N.E., E.W.), University of Melbourne, Australia
| |
Collapse
|
69
|
Zhou J, Zhang Z, Zhou H, Qian G. Diabetic Cognitive Dysfunction: From Bench to Clinic. Curr Med Chem 2020; 27:3151-3167. [PMID: 30727866 DOI: 10.2174/1871530319666190206225635] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 12/30/2018] [Accepted: 01/30/2019] [Indexed: 02/07/2023]
Abstract
Type 2 diabetes increases the risk of developing cognitive dysfunction in the elderly in the form of short-term memory and executive function impairment. Genetic and diet-induced models of type 2 diabetes further support this link, displaying deficits in working memory, learning, and memory performance. The risk factors for diabetic cognitive dysfunction include vascular disease, hypoglycaemia, hyperlipidaemia, adiposity, insulin resistance, lifestyle factors, and genetic factors. Using neuronal imaging technologies, diabetic patients with cognitive dysfunction show atrophy of the whole brain, particularly the grey matter, hippocampus and amygdala; increased volume of the ventricular and white matter; brain infarcts; impaired network integrity; abnormal microstructure; and reduced cerebral blood flow and amplitude of low-frequency fluctuations. The pathogenesis of type 2 diabetes with cognitive dysfunction involves hyperglycaemia, macrovascular and microvascular diseases, insulin resistance, inflammation, apoptosis, and disorders of neurotransmitters. Large clinical trials may offer further proof of biomarkers and risk factors for diabetic cognitive dysfunction. Advanced neuronal imaging technologies and novel disease animal models will assist in elucidating the precise pathogenesis and to provide better therapeutic interventions and treatment.
Collapse
Affiliation(s)
- Jiyin Zhou
- National Drug Clinical Trial Institution, the Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - Zuo Zhang
- National Drug Clinical Trial Institution, the Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - Hongli Zhou
- National Drug Clinical Trial Institution, the Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - Guisheng Qian
- Institute of Respiratory Diseases, the Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| |
Collapse
|
70
|
Cardiometabolic determinants of early and advanced brain alterations: Insights from conventional and novel MRI techniques. Neurosci Biobehav Rev 2020; 115:308-320. [DOI: 10.1016/j.neubiorev.2020.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/21/2020] [Accepted: 04/02/2020] [Indexed: 12/11/2022]
|
71
|
Söderbom G, Zeng BY. The NLRP3 inflammasome as a bridge between neuro-inflammation in metabolic and neurodegenerative diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 154:345-391. [PMID: 32739011 DOI: 10.1016/bs.irn.2020.03.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Evidence increasingly suggests that type 2 diabetes mellitus (T2DM) is a risk factor for neurodegenerative diseases (NDDs), such as Alzheimer's disease (AD) and Parkinson's disease (PD). These diseases share many pathological processes, including oxidative stress, local inflammation/neuroinflammation and chronic, low-grade (systemic) inflammation, which are exacerbated by aging, a common risk factor for T2DM and NDDs. Here, we focus on the link between chronic inflammation driven by peripheral metabolic disease and how this may impact neurodegeneration in AD and PD. We review the relationship between these common pathological processes in AD and PD from the perspective of the "pro-inflammatory" signaling of the nucleotide-binding oligomerization domain (NOD)-, leucine-rich repeat- (LRR)-, and pyrin domain-containing protein 3 (NLRP3) inflammasome complex. Since the need for effective disease-modifying therapies in T2DM, AD and PD is significant, the relationship between these diseases is important as a positive clinical impact on one may benefit the others. We briefly consider how novel strategies may target neuro-inflammation and provide potential therapies for AD and PD.
Collapse
Affiliation(s)
| | - Bai-Yun Zeng
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| |
Collapse
|
72
|
Du Y, Paiva K, Cebula A, Kim S, Lopez K, Li C, White C, Myneni S, Seshadri S, Wang J. Diabetes-Related Topics in an Online Forum for Caregivers of Individuals Living With Alzheimer Disease and Related Dementias: Qualitative Inquiry. J Med Internet Res 2020; 22:e17851. [PMID: 32628119 PMCID: PMC7381255 DOI: 10.2196/17851] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/07/2020] [Accepted: 06/03/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Diabetes and Alzheimer disease and related dementias (ADRD) are the seventh and sixth leading causes of death in the United States, respectively, and they coexist in many older adults. Caring for a loved one with both ADRD and diabetes is challenging and burdensome. OBJECTIVE This study aims to explore diabetes-related topics in the Alzheimer's Association ALZConnected caregiver forum by family caregivers of persons living with ADRD. METHODS User posts on the Alzheimer's Association ALZConnected caregiver forum were extracted. A total of 528 posts related to diabetes were included in the analysis. Of the users who generated the 528 posts, approximately 96.1% (275/286) were relatives of the care recipient with ADRD (eg, child, grandchild, spouse, sibling, or unspecified relative). Two researchers analyzed the data independently using thematic analysis. Any divergence was discussed among the research team, and an agreement was reached with a senior researcher's input as deemed necessary. RESULTS Thematic analysis revealed 7 key themes. The results showed that comorbidities of ADRD were common topics of discussions among family caregivers. Diabetes management in ADRD challenged family caregivers. Family caregivers might neglect their own health care because of the caring burden, and they reported poor health outcomes and reduced quality of life. The online forum provided a platform for family caregivers to seek support in their attempts to learn more about how to manage the ADRD of their care recipients and seek support for managing their own lives as caregivers. CONCLUSIONS The ALZConnected forum provided a platform for caregivers to seek informational and emotional support for caring for persons living with ADRD and diabetes. The overwhelming burdens with these two health conditions were apparent for both caregivers and care recipients based on discussions from the online forum. Studies are urgently needed to provide practical guidelines and interventions for diabetes management in individuals with diabetes and ADRD. Future studies to explore delivering diabetes management interventions through online communities in caregivers and their care recipients with ADRD and diabetes are warranted.
Collapse
Affiliation(s)
- Yan Du
- Center on Smart and Connected Health Technologies, School of Nursing, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Kristi Paiva
- Center on Smart and Connected Health Technologies, School of Nursing, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Adrian Cebula
- Center on Smart and Connected Health Technologies, School of Nursing, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Seon Kim
- Center on Smart and Connected Health Technologies, School of Nursing, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Katrina Lopez
- Center on Smart and Connected Health Technologies, School of Nursing, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Chengdong Li
- Center on Smart and Connected Health Technologies, School of Nursing, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Carole White
- School of Nursing, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Sahiti Myneni
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Jing Wang
- Center on Smart and Connected Health Technologies, School of Nursing, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
73
|
Stanciu GD, Bild V, Ababei DC, Rusu RN, Cobzaru A, Paduraru L, Bulea D. Link Between Diabetes and Alzheimer's Disease due to the Shared Amyloid Aggregation and Deposition Involving both Neurodegenerative Changes and Neurovascular Damages. J Clin Med 2020; 9:jcm9061713. [PMID: 32503113 PMCID: PMC7357086 DOI: 10.3390/jcm9061713] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/25/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023] Open
Abstract
Diabetes and Alzheimer’s disease are two highly prevalent diseases among the aging population and have become major public health concerns in the 21st century, with a significant risk to each other. Both of these diseases are increasingly recognized to be multifactorial conditions. The terms “diabetes type 3” or “brain diabetes” have been proposed in recent years to provide a complete view of the potential common pathogenic mechanisms between these diseases. While insulin resistance or deficiency remains the salient hallmarks of diabetes, cognitive decline and non-cognitive abnormalities such as impairments in visuospatial function, attention, cognitive flexibility, and psychomotor speed are also present. Furthermore, amyloid aggregation and deposition may also be drivers for diabetes pathology. Here, we offer a brief appraisal of social impact and economic burden of these chronic diseases and provide insight into amyloidogenesis through considering recent advances of amyloid-β aggregates on diabetes pathology and islet amyloid polypeptide on Alzheimer’s disease. Exploring the detailed knowledge of molecular interaction between these two amyloidogenic proteins opens new opportunities for therapies and biomarker development.
Collapse
Affiliation(s)
- Gabriela Dumitrita Stanciu
- Center for Advanced Research and Development in Experimental Medicine (CEMEX), Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania;
| | - Veronica Bild
- Center for Advanced Research and Development in Experimental Medicine (CEMEX), Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania;
- Pharmacodynamics and Clinical Pharmacy Department, Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (D.C.A.); (R.N.R.); (A.C.); (D.B.)
- Correspondence: (V.B.); (L.P.)
| | - Daniela Carmen Ababei
- Pharmacodynamics and Clinical Pharmacy Department, Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (D.C.A.); (R.N.R.); (A.C.); (D.B.)
| | - Razvan Nicolae Rusu
- Pharmacodynamics and Clinical Pharmacy Department, Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (D.C.A.); (R.N.R.); (A.C.); (D.B.)
| | - Alina Cobzaru
- Pharmacodynamics and Clinical Pharmacy Department, Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (D.C.A.); (R.N.R.); (A.C.); (D.B.)
| | - Luminita Paduraru
- Department Mother & Child Care, Division Neonatology, Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
- Correspondence: (V.B.); (L.P.)
| | - Delia Bulea
- Pharmacodynamics and Clinical Pharmacy Department, Grigore T. Popa” University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania; (D.C.A.); (R.N.R.); (A.C.); (D.B.)
| |
Collapse
|
74
|
Abstract
Tau protein which was discovered in 1975 [310] became of great interest when it was identified as the main component of neurofibrillary tangles (NFT), a pathological feature in the brain of patients with Alzheimer's disease (AD) [39, 110, 232]. Tau protein is expressed mainly in the brain as six isoforms generated by alternative splicing [46, 97]. Tau is a microtubule associated proteins (MAPs) and plays a role in microtubules assembly and stability, as well as diverse cellular processes such as cell morphogenesis, cell division, and intracellular trafficking [49]. Additionally, Tau is involved in much larger neuronal functions particularly at the level of synapses and nuclei [11, 133, 280]. Tau is also physiologically released by neurons [233] even if the natural function of extracellular Tau remains to be uncovered (see other chapters of the present book).
Collapse
|
75
|
Yang H, Hong W, Chen L, Tao Y, Peng Z, Zhou H. Analysis of risk factors for depression in Alzheimer's disease patients. Int J Neurosci 2020; 130:1136-1141. [PMID: 32053409 DOI: 10.1080/00207454.2020.1730369] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Purpose: Depression, which affects about 52% of Alzheimer's disease (AD) patients, can worsen cognitive impairment and increase mortality and suicide rates. We hope to provide clinical evidence for the prevention and treatment of depression in AD patients by investigating related risk factors of depression in AD patients.Methods: 158 AD inpatients of the Department of Neurology, Daping Hospital from September 2017 to March 2019 were enrolled. General information, laboratory tests, cognitive and emotional function assessments of the inpatients were collected. Logistic regression was used to analyze the risk factors of depression in AD patients, and the relationship between 17 Hamilton depression scale scores and HbA1c levels in AD patients was further analyzed.Results: The prevalence of age, gender, hypertension, hyperlipidemia, Type 2 diabetes mellitus (T2DM), and white matter lesions (WML) in the AD with depression group was significantly different from without depression group. Hypertension, T2DM, and WML are independent risk factors for depression in AD patients. The depression scores of AD patients with HbA1c>6.5% were significantly higher than AD patients with HbA1c ≤ 6.5%, and there were significant difference in depression scale scores between using anti-diabetes drugs group and not using anti-diabetes drugs group whose HbA1c level is >6.5%, while no difference in depression scores between using anti-diabetes drugs group and not using anti-diabetes drugs group whose HbA1c level is ≤6.5%.Conclusion: T2DM is an independent risk factor for AD patients with depression. Increased HbA1c levels aggravate depression in AD patients, and controlling HbA1c levels and anti-diabetes drugs can reduce the severity of depression in AD patients.
Collapse
Affiliation(s)
- Hai Yang
- Department of Neurology, Daping Hospital, Army Military Medical University, Chongqing, China
| | - Wenjuan Hong
- Graduate School of Bengbu Medical College, Bengbu City, China
| | - Le Chen
- Graduate School of Bengbu Medical College, Bengbu City, China
| | - Yong Tao
- Department of Neurology, Daping Hospital, Army Military Medical University, Chongqing, China
| | - Zeyan Peng
- Department of Neurology, Daping Hospital, Army Military Medical University, Chongqing, China
| | - Huadong Zhou
- Department of Neurology, Daping Hospital, Army Military Medical University, Chongqing, China
| |
Collapse
|
76
|
Faraone I, Russo D, Chiummiento L, Fernandez E, Choudhary A, Monné M, Milella L, Rai DK. Phytochemicals of Minthostachys diffusa Epling and Their Health-Promoting Bioactivities. Foods 2020; 9:foods9020144. [PMID: 32024045 PMCID: PMC7074199 DOI: 10.3390/foods9020144] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 12/31/2022] Open
Abstract
The genus Minthostachys belonging to the Lamiaceae family, and is an important South American mint genus used commonly in folk medicine as an aroma in cooking. The phytochemical-rich samples of the aerial parts of Minthostachys diffusa Epling. were tested for pharmacological and health-promoting bioactivities using in vitro chemical and enzymatic assays. A range of radical scavenging activities of the samples against biological radicals such as nitric oxide and superoxide anion and against synthetic 2,2-diphenyl-1-picrylhydrazyl and 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) radicals, the ferric reducing antioxidant power and the lipid peroxidation inhibition were determined and ranked using the ‘relative antioxidant capacity index’ (RACI). The ethyl acetate fraction showed the highest RACI of +1.12. Analysis of the various fractions’ inhibitory ability against enzymes involved in diabetes (α-amylase and α-glucosidase), and against enzymes associated with Parkinson’s or Alzheimer’s diseases (acetylcholinesterase and butyrylcholinesterase) also suggested that the ethyl acetate fraction was the most active. Liquid chromatography–tandem mass spectrometry analysis of the ethyl acetate fraction showed more than 30 polyphenolic compounds, including triterpenes. The inhibitory cholinesterase effects of the triterpenes identified from M. diffusa were further analysed by in silico docking of these compounds into 3D-structures of acetylcholinesterase and butyrylcholinesterase. This is the first study on pharmacological activities and phytochemical profiling of the aerial parts of M. diffusa, showing that this plant, normally used as food in South America, is also rich in health-promoting phytochemicals.
Collapse
Affiliation(s)
- Immacolata Faraone
- Department of Science, University of Basilicata, V.le dell’Ateneo Lucano, 10, 85100 Potenza, Italy; (I.F.); (D.R.); (L.C.); (M.M.)
- Spinoff BioActiPlant s.r.l., Università della Basilicata, V.le dell’Ateneo Lucano, 10, 85100 Potenza, Italy
| | - Daniela Russo
- Department of Science, University of Basilicata, V.le dell’Ateneo Lucano, 10, 85100 Potenza, Italy; (I.F.); (D.R.); (L.C.); (M.M.)
- Spinoff BioActiPlant s.r.l., Università della Basilicata, V.le dell’Ateneo Lucano, 10, 85100 Potenza, Italy
| | - Lucia Chiummiento
- Department of Science, University of Basilicata, V.le dell’Ateneo Lucano, 10, 85100 Potenza, Italy; (I.F.); (D.R.); (L.C.); (M.M.)
| | - Eloy Fernandez
- Department of Crop Sciences and Agroforestry, Faculty of Tropical AgriSciences, Czech University of Life Sciences, Praha 6-Suchdol, Kamýcká 129, 16500 Prague, Czech Republic;
| | - Alka Choudhary
- Department of Food BioSciences, Teagasc Food Research Centre Ashtown, D15KN3K Dublin, Ireland; (A.C.); (D.K.R.)
| | - Magnus Monné
- Department of Science, University of Basilicata, V.le dell’Ateneo Lucano, 10, 85100 Potenza, Italy; (I.F.); (D.R.); (L.C.); (M.M.)
| | - Luigi Milella
- Department of Science, University of Basilicata, V.le dell’Ateneo Lucano, 10, 85100 Potenza, Italy; (I.F.); (D.R.); (L.C.); (M.M.)
- Spinoff BioActiPlant s.r.l., Università della Basilicata, V.le dell’Ateneo Lucano, 10, 85100 Potenza, Italy
- Correspondence: ; Tel./Fax: +39-0971-205525
| | - Dilip K. Rai
- Department of Food BioSciences, Teagasc Food Research Centre Ashtown, D15KN3K Dublin, Ireland; (A.C.); (D.K.R.)
| |
Collapse
|
77
|
Esmaeili MH, Enayati M, Khabbaz Abkenar F, Ebrahimian F, Salari AA. Glibenclamide mitigates cognitive impairment and hippocampal neuroinflammation in rats with type 2 diabetes and sporadic Alzheimer-like disease. Behav Brain Res 2020; 379:112359. [PMID: 31733313 DOI: 10.1016/j.bbr.2019.112359] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 10/23/2019] [Accepted: 11/13/2019] [Indexed: 12/20/2022]
|
78
|
Diabetes mellitus in the young and the old: Effects on cognitive functioning across the life span. Neurobiol Dis 2020; 134:104608. [DOI: 10.1016/j.nbd.2019.104608] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 08/06/2019] [Accepted: 09/04/2019] [Indexed: 01/12/2023] Open
|
79
|
Association between diabetes and cognitive function at baseline in the Brazilian Longitudinal Study of Adult Health (ELSA- Brasil). Sci Rep 2020; 10:1596. [PMID: 32005901 PMCID: PMC6994611 DOI: 10.1038/s41598-020-58332-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 01/06/2020] [Indexed: 01/21/2023] Open
Abstract
Diabetes has been associated with cognitive changes and an increased risk of vascular dementia and Alzheimer’s disease, but it is unclear whether there are associations between diabetes and early alterations in cognitive performance. The present study consisted of a cross-section analysis of 14,444 participants aged 35–74 years and from a developing country at baseline in the Brazilian Longitudinal Study of Adult Health (ELSA–Brasil); these participants were recruited between 2008 and 2010. We investigated whether there was an association between diabetes and early changes in the cognitive performance of this Brazilian population. To assess cognitive domains, we used the word-list learning, word-list delayed recall and word recognition tests along. Phonemic verbal fluency tests included semantic phonemic test (animals) and a phonemic test (words beginning with the letter F). Executive functions associated with attention, concentration and psychomotor speed were evaluated using the Trail Making Test B. The exposure variable in the study was defined as diabetes. Multiple linear regression was used to estimate the association between diabetes and cognitive performance. The results were adjusted for age, sex, education, hypertension, coronary disease, depression, physical activity, smoking, alcohol consumption, and the cholesterol/HDL-C ratio. We found a significant association between diabetes and decreased memory, language and executive function (attention, concentration and psychomotor speed) performance in this population from a country with a distinct epidemiological profile, even after adjusting for the main intervening variables.
Collapse
|
80
|
Shi Q, Zhou F, Mei J, Yang H, Li H. The Effect of Type 2 Diabetes Mellitus on Neuropsychological Symptoms in Chinese Early Alzheimer's Disease Population. Neuropsychiatr Dis Treat 2020; 16:829-836. [PMID: 32273709 PMCID: PMC7105356 DOI: 10.2147/ndt.s240529] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 03/01/2020] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE To explore the effect of Type 2 diabetes mellitus (T2DM) on the development of neuropsychiatric symptoms (NPS) in early Alzheimer's disease (AD). METHODS From September 2017 to March 2019, a cross-sectional study was conducted on the clinical data of 158 early AD patients over 65 years old in the Department of Neurology of Daping Hospital. All early stage of AD patients were divided into early stage of AD with NPS group and early stage of AD without NPS group according to the presence or absence of NPS. Clinical data of age, sex, body mass index (BMI), smoking and alcohol consumption, history of hypertension, hyperlipidemia, white matter leisure (WML) and T2DM, MMSE, CDR and NPI-Q scores were collected. Multivariate logistic regression analyses were performed to examine the relationship between T2DM and NPS in early AD. RESULTS Compared with the early stage of AD group without NPS, the early stage of AD group with NPS was older, the proportion of women was higher, the proportion of T2DM, hypertension, hyperlipidemia and WML was higher, and the MMSE score was lower (P< 0.05). T2DM was an independent risk factor for NPS in early stage of AD patients (OR 3.48, 95% CI 2.91-3.84). The incidence of T2DM in AD patients with depression, anxiety, nighttime behavioral disturbances, and appetite disturbances was significantly higher than in AD patients without these symptoms. T2DM was an independent risk factor of depression (OR 2.04, 95% CI 1.71-2.38), anxiety (OR 1.69, 95% CI 1.38-1.97), nighttime behavioral disturbances (OR 1.95, 95% CI 1.75-2.13) and appetite disturbances (OR 1.62, 95% CI 1.33-1.94) in early AD patients. CONCLUSION T2DM was an important independent risk factor for NPS in early AD, which promotes the occurrence of depression, anxiety, nighttime behavioral disturbances and appetite disturbances in early AD.
Collapse
Affiliation(s)
- Qianqian Shi
- Department of Neurology, Daping Hospital, Army Medical University, Chongqing 400042, People's Republic of China
| | - Faying Zhou
- Department of Neurology, Daping Hospital, Army Medical University, Chongqing 400042, People's Republic of China
| | - Jing Mei
- Department of Neurology, Daping Hospital, Army Medical University, Chongqing 400042, People's Republic of China
| | - Haimei Yang
- Department of Neurology, Daping Hospital, Army Medical University, Chongqing 400042, People's Republic of China
| | - Huiyun Li
- Department of Neurology, Daping Hospital, Army Medical University, Chongqing 400042, People's Republic of China
| |
Collapse
|
81
|
Abstract
The American Diabetes Association (ADA) "Standards of Medical Care in Diabetes" includes the 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 (https://doi.org/10.2337/dc20-SPPC), 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 (https://doi.org/10.2337/dc20-SINT). Readers who wish to comment on the Standards of Care are invited to do so at professional.diabetes.org/SOC.
Collapse
|
82
|
Kim J, Park E, An M. The Cognitive Impact of Chronic Diseases on Functional Capacity in Community-Dwelling Adults. J Nurs Res 2019; 27:1-8. [PMID: 29985821 PMCID: PMC6369881 DOI: 10.1097/jnr.0000000000000272] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Background: People with chronic diseases may experience poor cognitive functioning associated with advanced age, progression of disease, or other comorbid chronic conditions. Empirical evidence of this phenomenon is limited despite the clinical relevance of cognitive decline and associated adverse outcomes such as poor physical functioning. Purpose: The purpose of this study was to examine cognitive functioning in the domains of memory, attention, and executive function and its association with functional capacity in a sample of community-dwelling adults with a spectrum of chronic diseases. Methods: An exploratory cross-sectional study was conducted in a sample of community-dwelling adults with chronic diseases, including hypertension (58.9%), diabetes mellitus (DM; 20.0%), and dyslipidemia (14.4%). Participants’ mean age was 64.1 ± 11.2 years, and 48.9% were male. Ninety persons completed the face-to-face interviews, which evaluated cognitive functioning in the domains of memory, attention, and executive function using neuropsychological tests and the physical well-being test, which measured functional capacity using the Duke Activity Status Index. Results: Compared with those with other chronic diseases, our sample with hypertension and DM had significantly more memory loss and poorer executive function. These significant differences were nullified when adjusting for age, gender, and education. Approximately one third had functional limitations (n = 29, 32.2%), using a cutoff point of 35 or less (Duke Activity Status Index). Memory loss (delayed recall, b = 1.5, p = .016) and poor executive function (Trail Making Test Part A, b = −0.2, p < .001) were predicting factors of functional decline, independent of age, gender, education, and comorbidity. Conclusions/Implications for Practice: Cognitive function, particularly memory and executive function, was poorer among chronically ill Korean adults in the community with hypertension or DM than their counterparts. Functional decline was worse in the presence of memory loss and poor executive function. Studies examining the mechanism by which overall functioning is impacted by cognitive decline and its relevance to functional declines in a larger representative sample are warranted.
Collapse
Affiliation(s)
- JinShil Kim
- PhD, RN, Professor, College of Nursing, Gachon University, Incheon, South Korea
| | | | - Minjeong An
- PhD, RN, Assistant Professor, College of Nursing, Chonnam National University, Gwangju, South Korea
| |
Collapse
|
83
|
Oxidized cholesterol species as signaling molecules in the brain: diabetes and Alzheimer's disease. Neuronal Signal 2019; 3:NS20190068. [PMID: 32269839 PMCID: PMC7104322 DOI: 10.1042/ns20190068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 01/16/2023] Open
Abstract
Type 2 diabetes is associated with adverse central nervous system effects, including a doubled risk for Alzheimer's disease (AD) and increased risk of cognitive impairment, but the mechanisms connecting diabetes to cognitive decline and dementia are unknown. One possible link between these diseases may be the associated alterations to cholesterol oxidation and metabolism in the brain. We will survey evidence demonstrating alterations to oxysterols in the brain in AD and diabetes and how these oxysterols could contribute to pathology, as well as identifying research questions that have not yet been addressed to allow for a fuller understanding of the role of oxysterols in AD and diabetes.
Collapse
|
84
|
Crook JE, Gunter JL, Ball CT, Jones DT, Graff-Radford J, Knopman DS, Boeve BF, Petersen RC, Jack CR, Graff-Radford NR. Linear vs volume measures of ventricle size: Relation to present and future gait and cognition. Neurology 2019; 94:e549-e556. [PMID: 31748251 DOI: 10.1212/wnl.0000000000008673] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 07/31/2019] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE To compare the clinical utility of volume-based ratios with the standard linear ratio of Evans index (EI) by examining their associations with gait, cognition, and other patient and imaging variables. METHODS From MRI scans of 1,774 participants in the Mayo Clinic Study of Aging, we calculated 3 ventricle size measures: Evan index (frontal horn width divided by widest width of skull inner table), total ventricular volume, and frontal horn volume as ratios of total intracranial volume. Gait was measured by a timed 25-foot walk and cognition by a composite of psychometric tests. We also evaluated variables associated with the measures of ventricular size. Further, we evaluated gait and cognition associations with MRI of extraventricular findings seen in normal-pressure hydrocephalus: disproportionate enlargement of subarachnoid space (DESH) and focal sulcal dilations (FSD). RESULTS Ventricular volume measures had stronger association with gait and cognition measures than EI. In decreasing order of strength of association with ventricle size were DESH, FSD, white matter hyperintensity volume ratio, age, male sex, cortical thickness, and education. Modest evidence was observed that FSD was associated with future decline in gait and cognition. CONCLUSION Ventricular volume measures are clinically more useful than EI in indicating current and future gait and cognition. Multiple factors are associated with ventricle volume size, including FSD and DESH, suggesting that changes in CSF dynamics may go beyond simple ventriculomegaly.
Collapse
Affiliation(s)
- Julia E Crook
- From the Departments of Health Sciences Research (J.E.C., C.T.B.) and Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; and Departments of Radiology (J.L.G., C.R.J.) and Neurology (D.T.J., J.G.-R., D.S.K., B.F.B., R.C.P.), Mayo Clinic, Rochester, MN
| | - Jeffrey L Gunter
- From the Departments of Health Sciences Research (J.E.C., C.T.B.) and Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; and Departments of Radiology (J.L.G., C.R.J.) and Neurology (D.T.J., J.G.-R., D.S.K., B.F.B., R.C.P.), Mayo Clinic, Rochester, MN
| | - Colleen T Ball
- From the Departments of Health Sciences Research (J.E.C., C.T.B.) and Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; and Departments of Radiology (J.L.G., C.R.J.) and Neurology (D.T.J., J.G.-R., D.S.K., B.F.B., R.C.P.), Mayo Clinic, Rochester, MN
| | - David T Jones
- From the Departments of Health Sciences Research (J.E.C., C.T.B.) and Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; and Departments of Radiology (J.L.G., C.R.J.) and Neurology (D.T.J., J.G.-R., D.S.K., B.F.B., R.C.P.), Mayo Clinic, Rochester, MN
| | - Jonathan Graff-Radford
- From the Departments of Health Sciences Research (J.E.C., C.T.B.) and Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; and Departments of Radiology (J.L.G., C.R.J.) and Neurology (D.T.J., J.G.-R., D.S.K., B.F.B., R.C.P.), Mayo Clinic, Rochester, MN
| | - David S Knopman
- From the Departments of Health Sciences Research (J.E.C., C.T.B.) and Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; and Departments of Radiology (J.L.G., C.R.J.) and Neurology (D.T.J., J.G.-R., D.S.K., B.F.B., R.C.P.), Mayo Clinic, Rochester, MN
| | - Bradley F Boeve
- From the Departments of Health Sciences Research (J.E.C., C.T.B.) and Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; and Departments of Radiology (J.L.G., C.R.J.) and Neurology (D.T.J., J.G.-R., D.S.K., B.F.B., R.C.P.), Mayo Clinic, Rochester, MN
| | - Ronald C Petersen
- From the Departments of Health Sciences Research (J.E.C., C.T.B.) and Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; and Departments of Radiology (J.L.G., C.R.J.) and Neurology (D.T.J., J.G.-R., D.S.K., B.F.B., R.C.P.), Mayo Clinic, Rochester, MN
| | - Clifford R Jack
- From the Departments of Health Sciences Research (J.E.C., C.T.B.) and Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; and Departments of Radiology (J.L.G., C.R.J.) and Neurology (D.T.J., J.G.-R., D.S.K., B.F.B., R.C.P.), Mayo Clinic, Rochester, MN
| | - Neill R Graff-Radford
- From the Departments of Health Sciences Research (J.E.C., C.T.B.) and Neurology (N.R.G.-R.), Mayo Clinic, Jacksonville, FL; and Departments of Radiology (J.L.G., C.R.J.) and Neurology (D.T.J., J.G.-R., D.S.K., B.F.B., R.C.P.), Mayo Clinic, Rochester, MN.
| |
Collapse
|
85
|
Sanjari Moghaddam H, Ghazi Sherbaf F, Aarabi MH. Brain microstructural abnormalities in type 2 diabetes mellitus: A systematic review of diffusion tensor imaging studies. Front Neuroendocrinol 2019; 55:100782. [PMID: 31401292 DOI: 10.1016/j.yfrne.2019.100782] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 07/27/2019] [Accepted: 08/07/2019] [Indexed: 12/13/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is associated with deficits in the structure and function of the brain. Diffusion tensor imaging (DTI) is a highly sensitive method for characterizing cerebral tissue microstructure. Using PRISMA guidelines, we identified 29 studies which have demonstrated widespread brain microstructural impairment and topological network disorganization in patients with T2DM. Most consistently reported structures with microstructural abnormalities were frontal, temporal, and parietal lobes in the lobar cluster; corpus callosum, cingulum, uncinate fasciculus, corona radiata, and internal and external capsules in the white matter cluster; thalamus in the subcortical cluster; and cerebellum. Microstructural abnormalities were correlated with pathological derangements in the endocrine profile as well as deficits in cognitive performance in the domains of memory, information-processing speed, executive function, and attention. Altogether, the findings suggest that the detrimental effects of T2DM on cognitive functions might be due to microstructural disruptions in the central neural structures.
Collapse
Affiliation(s)
| | - Farzaneh Ghazi Sherbaf
- Neuroradiology Division, Tehran University of Medical Sciences, School of Medicine, Tehran, Iran
| | - Mohammad Hadi Aarabi
- Neuroradiology Division, Tehran University of Medical Sciences, School of Medicine, Tehran, Iran.
| |
Collapse
|
86
|
Bae IS, Kim JM, Cheong JH, Ryu JI, Han MH. Association between bone mineral density and brain parenchymal atrophy and ventricular enlargement in healthy individuals. Aging (Albany NY) 2019; 11:8217-8238. [PMID: 31575827 PMCID: PMC6814624 DOI: 10.18632/aging.102316] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 09/21/2019] [Indexed: 01/01/2023]
Abstract
Bone, vascular smooth muscle, and arachnoid trabeculae are composed of the same type of collagen. However, no studies have investigated the relationship between bone mineral density deterioration and cerebral atrophy, both of which occur in normal, healthy aging. Accordingly, we evaluated whether bone mineral density was associated with brain parenchymal atrophy and ventricular enlargement in healthy individuals. Intracranial cavity, brain parenchyma, and lateral ventricles volumes were measured using brain magnetic resonance imaging (MRI) with a semiautomated tool. We included 267 individuals with no history of dementia or other neurological diseases, who underwent one or more dual-energy X-ray absorptiometry scans and brain MRIs simultaneously (within 3 years of each other) at our hospital over an 11-year period. We found that progression of brain parenchymal atrophy was positively associated with bone mineral density after full adjustment (B, 0.94; P < 0.001). In addition, individuals with osteoporosis showed more parenchymal atrophy among those younger than 80 years. In addition, we observed greater ventricular enlargement in individuals with osteoporosis among those older than 80 years. We believe that osteoporosis may play a role in the acceleration of parenchymal atrophy during the early-stages, and ventricular enlargement in the late-stages, of normal aging-related cerebral atrophy.
Collapse
Affiliation(s)
- In-Suk Bae
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri, Gyonggi-do, Korea
| | - Jae Min Kim
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri, Gyonggi-do, Korea
| | - Jin Hwan Cheong
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri, Gyonggi-do, Korea
| | - Je Il Ryu
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri, Gyonggi-do, Korea
| | - Myung-Hoon Han
- Department of Neurosurgery, Hanyang University Guri Hospital, Guri, Gyonggi-do, Korea
| |
Collapse
|
87
|
Chatterjee S, Ambegaokar SS, Jackson GR, Mudher A. Insulin-Mediated Changes in Tau Hyperphosphorylation and Autophagy in a Drosophila Model of Tauopathy and Neuroblastoma Cells. Front Neurosci 2019; 13:801. [PMID: 31427921 PMCID: PMC6688711 DOI: 10.3389/fnins.2019.00801] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/17/2019] [Indexed: 12/13/2022] Open
Abstract
Almost 50 million people in the world are affected by dementia; the most prevalent form of which is Alzheimer’s disease (AD). Although aging is considered to be the main risk factor for AD, growing evidence from epidemiological studies suggests that type 2 diabetes mellitus (T2DM) increases the risk of dementia including AD. Defective brain insulin signaling has been suggested as an early event in AD and other tauopathies but the mechanisms that link these diseases are largely unknown. Tau hyperphosphorylation is a hallmark of neurofibrillary pathology and insulin resistance increases the number of neuritic plaques particularly in AD. Utilizing a combination of our Drosophila models of tauopathy (expressing the 2N4R-Tau) and neuroblastoma cells, we have attempted to decipher the pathways downstream of the insulin signaling cascade that lead to tau hyperphosphorylation, aggregation and autophagic defects. Using cell-based, genetic, and biochemical approaches we have demonstrated that tau phosphorylation at AT8 and PHF1 residues is enhanced in an insulin-resistant environment. We also show that insulin-induced changes in total and phospho-tau are mediated by the crosstalk of AKT, glycogen synthase kinase-3β, and extracellular regulating kinase located downstream of the insulin receptor pathway. Finally, we demonstrate a significant change in the levels of the key proteins in the mammalian target of rapamycin/autophagy pathway, implying an increased impairment of aggregated protein clearance in our transgenic Drosophila models and cultured neuroblastoma cells.
Collapse
Affiliation(s)
- Shreyasi Chatterjee
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch at Galveston, Galveston, TX, United States.,Department of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Suren S Ambegaokar
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch at Galveston, Galveston, TX, United States.,Department of Botany and Microbiology, Ohio Wesleyan University, Delaware, OH, United States
| | - George R Jackson
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch at Galveston, Galveston, TX, United States.,Department of Neurology, Michael E. DeBakey VA Medical Center, Parkinson's Disease Research Education and Clinical Center, Baylor College of Medicine, Houston, TX, United States
| | - Amritpal Mudher
- Department of Biological Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
88
|
Dong S, Dongwei L, Zhang J, Liang J, Sun Z, Fang J. Individuals in the prediabetes stage exhibit reduced hippocampal tail volume and executive dysfunction. Brain Behav 2019; 9:e01351. [PMID: 31240857 PMCID: PMC6710206 DOI: 10.1002/brb3.1351] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 05/05/2019] [Accepted: 05/28/2019] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION High glucose levels are associated with cognitive impairment and total hippocampal volume reductions. However, the effects of the blood glucose level on hippocampal subfield volumes remain unclear, especially in the prediabetes stage. METHODS Sixty participants were enrolled in this cross-sectional study and were divided into the nondiabetes, prediabetes, and diabetes groups according to their medical history and A1c level. A full battery of neuropsychological tests was used to assess the global cognition, executive function, attention, verbal fluency, working memory, immediate memory, and delayed memory. FreeSurfer 6.0 was used for the hippocampus parcellation. Hippocampal subfield volumes were adjusted by intracranial volume. Analyses of covariance, multiple linear regression, and partial correlation analysis were used to explore the relationship between A1c level, cognitive function, and hippocampal subfields volume, in which age, sex, education years, body mass index, history of hypertension, level of cholesterol, and the presence of ApoE4-positive status were adjusted. RESULTS Significant differences in the total left hippocampal volume (p = 0.046) and left hippocampal tail volume (p = 0.014) were noted among three groups. Significant correlation was identified between the A1c level and the volume of left hippocampal tail (r = -0.334, p = 0.009) after adjusting all the covariants. Increased A1c level was significantly associated with executive dysfunction, as assessed by trail making test B (R = 0.503, p = 0.0016) and Stroop test C (R = 0.506, p = 0.001). CONCLUSIONS Our results support that the left hippocampal tail volume may be served as an early marker of diabetes-related brain damage, associated with executive dysfunction. Clinicians should pay closer attention to adults in the prediabetes stage to prevent later cognitive impairment.
Collapse
Affiliation(s)
- Sun Dong
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lu Dongwei
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Junjian Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jinyu Liang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhenmeng Sun
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jian Fang
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
89
|
Cholerton B, Omidpanah A, Verney SP, Nelson LA, Baker LD, Suchy-Dicey A, Longstreth WT, Howard BV, Henderson JA, Montine TJ, Buchwald D. Type 2 diabetes and later cognitive function in older American Indians: The Strong Heart Study. Int J Geriatr Psychiatry 2019; 34:1050-1057. [PMID: 30924200 PMCID: PMC6579638 DOI: 10.1002/gps.5108] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 11/29/2018] [Indexed: 01/17/2023]
Abstract
OBJECTIVES Insulin resistance is a substantial health issue for American Indians, with type 2 diabetes overrepresented in this population as compared with non-Hispanic whites. Insulin resistance and its related conditions in turn increase risk for dementia and cognitive impairment. The aim of the current study was to determine whether type 2 diabetes and insulin resistance at midlife was associated with later-life cognitive testing in a large sample of older American Indians, aged 65 and older. METHODS American Indian participants who underwent both fasting blood draw as part of the Strong Heart Study and had subsequent cognitive testing as part of the later adjunct Cerebrovascular Disease and its Consequences in American Indians study were included (n = 790). Regression models examined type 2 diabetes and impaired fasting glucose and subsequent cognitive test performance as part of a longitudinal study design. The relationship between a continuous measure of insulin resistance and later cognitive test performance was assessed using generalized estimating equations. RESULTS Controlling for demographic and clinical factors, verbal fluency and processing speed/working memory were significantly negatively associated with having type 2 diabetes and with insulin resistance, but not with impaired fasting glucose. CONCLUSION In this sample of American Indians, type 2 diabetes at midlife was associated with subsequent lower performance on measures of executive function. These results may have important implications for future implementation of diagnostic and intervention services in this population.
Collapse
Affiliation(s)
- Brenna Cholerton
- Department of Pathology, Stanford University, Palo Alto, California, USA
| | - Adam Omidpanah
- Department of Community Health, Washington State University, Seattle, Washington, USA
| | - Steven P. Verney
- Department of Psychology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Lonnie A. Nelson
- Department of Community Health, Washington State University, Seattle, Washington, USA
| | - Laura D. Baker
- Department of Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Astrid Suchy-Dicey
- Department of Community Health, Washington State University, Seattle, Washington, USA
| | - William T. Longstreth
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington, USA,Department of Epidemiology, School of Public Health, University of Washington, Seattle, Washington, USA
| | | | | | - Thomas J. Montine
- Department of Pathology, Stanford University, Palo Alto, California, USA
| | - Dedra Buchwald
- Department of Community Health, Washington State University, Seattle, Washington, USA
| |
Collapse
|
90
|
Ryan L, Hay M, Huentelman MJ, Duarte A, Rundek T, Levin B, Soldan A, Pettigrew C, Mehl MR, Barnes CA. Precision Aging: Applying Precision Medicine to the Field of Cognitive Aging. Front Aging Neurosci 2019; 11:128. [PMID: 31231204 PMCID: PMC6568195 DOI: 10.3389/fnagi.2019.00128] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 05/16/2019] [Indexed: 12/13/2022] Open
Abstract
The current "one size fits all" approach to our cognitive aging population is not adequate to close the gap between cognitive health span and lifespan. In this review article, we present a novel model for understanding, preventing, and treating age-related cognitive impairment (ARCI) based on concepts borrowed from precision medicine. We will discuss how multiple risk factors can be classified into risk categories because of their interrelatedness in real life, the genetic variants that increase sensitivity to, or ameliorate, risk for ARCI, and the brain drivers or common mechanisms mediating brain aging. Rather than providing a definitive model of risk for ARCI and cognitive decline, the Precision Aging model is meant as a starting point to guide future research. To that end, after briefly discussing key risk categories, genetic risks, and brain drivers, we conclude with a discussion of steps that must be taken to move the field forward.
Collapse
Affiliation(s)
- Lee Ryan
- Department of Psychology, College of Science, University of Arizona, Tucson, AZ, United States
| | - Meredith Hay
- Department of Physiology, University of Arizona, Tucson, AZ, United States
| | - Matt J. Huentelman
- Neurobehavioral Research Unit, Division of Neurological Disorders, Translational Genomics Research Institute (TGen), Phoenix, AZ, United States
| | - Audrey Duarte
- Center for Advanced Brain Imaging, School of Psychology, Georgia Institute of Technology, Atlanta, GA, United States
| | - Tatjana Rundek
- Clinical and Translational Research Division, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Bonnie Levin
- Neuropsychology Division, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Anja Soldan
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Corinne Pettigrew
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Matthias R. Mehl
- Department of Psychology, College of Science, University of Arizona, Tucson, AZ, United States
| | - Carol A. Barnes
- Department of Psychology, College of Science, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
91
|
Boots EA, Zhan L, Dion C, Karstens AJ, Peven JC, Ajilore O, Lamar M. Cardiovascular disease risk factors, tract-based structural connectomics, and cognition in older adults. Neuroimage 2019; 196:152-160. [PMID: 30980900 DOI: 10.1016/j.neuroimage.2019.04.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 03/29/2019] [Accepted: 04/05/2019] [Indexed: 01/01/2023] Open
Abstract
Cardiovascular disease risk factors (CVD-RFs) are associated with decreased gray and white matter integrity and cognitive impairment in older adults. Less is known regarding the interplay between CVD-RFs, brain structural connectome integrity, and cognition. We examined whether CVD-RFs were associated with measures of tract-based structural connectivity in 94 non-demented/non-depressed older adults and if alterations in connectivity mediated associations between CVD-RFs and cognition. Participants (age = 68.2 years; 52.1% female; 46.8% Black) underwent CVD-RF assessment, MRI, and cognitive evaluation. Framingham 10-year stroke risk (FSRP-10) quantified CVD-RFs. Graph theory analysis integrated T1-derived gray matter regions of interest (ROIs; 23 a-priori ROIs associated with CVD-RFs and dementia), and diffusion MRI-derived white matter tractography into connectivity matrices analyzed for local efficiency and nodal strength. A principal component analysis resulted in three rotated factor scores reflecting executive function (EF; FAS, Trail Making Test (TMT) B-A, Letter-Number Sequencing, Matrix Reasoning); attention/information processing (AIP; TMT-A, TMT-Motor, Digit Symbol); and memory (CVLT-II Trials 1-5 Total, Delayed Free Recall, Recognition Discriminability). Linear regressions between FSRP-10 and connectome ROIs adjusting for word reading, intracranial volume, and white matter hyperintensities revealed negative associations with nodal strength in eight ROIs (p-values<.05) and negative associations with efficiency in two ROIs, and a positive association in one ROI (p-values<.05). There was mediation of bilateral hippocampal strength on FSRP-10 and AIP, and left rostral middle frontal gyrus strength on FSRP-10 and AIP and EF. Stroke risk plays differential roles in connectivity and cognition, suggesting the importance of multi-modal neuroimaging biomarkers in understanding age-related CVD-RF burden and brain-behavior.
Collapse
Affiliation(s)
- Elizabeth A Boots
- Department of Psychology, University of Illinois at Chicago, Chicago, IL, 60607, USA; Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Liang Zhan
- Department of Electrical and Computer Engineering, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Catherine Dion
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, 32603, USA
| | - Aimee J Karstens
- Department of Psychology, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Jamie C Peven
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Olusola Ajilore
- Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Melissa Lamar
- Department of Psychology, University of Illinois at Chicago, Chicago, IL, 60607, USA; Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA; Department of Behavioral Sciences, Rush University Medical Center, Chicago, IL, 60612, USA.
| |
Collapse
|
92
|
Callisaya ML, Beare R, Moran C, Phan T, Wang W, Srikanth VK. Type 2 diabetes mellitus, brain atrophy and cognitive decline in older people: a longitudinal study. Diabetologia 2019; 62:448-458. [PMID: 30547230 DOI: 10.1007/s00125-018-4778-9] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 10/22/2018] [Indexed: 12/14/2022]
Abstract
AIMS/HYPOTHESIS The aims of the study were to examine whether type 2 diabetes mellitus is associated with greater brain atrophy and cognitive decline, and whether brain atrophy mediates associations between type 2 diabetes and cognitive decline. METHODS Participants without dementia aged 55-90 years from the Cognition and Diabetes in Older Tasmanians (CDOT) study underwent brain MRI (ventricular and total brain volume) and neuropsychological measures (global function and seven cognitive domains) at three time points over 4.6 years. Mixed models were used to examine longitudinal associations of type 2 diabetes with cognitive and MRI measures, adjusting for covariates. A test of mediation was used to determine whether brain atrophy explained associations between type 2 diabetes and cognitive decline. RESULTS A total of 705 participants (diabetes: n = 348, mean age 68.2 years [SD 7.0]; no diabetes: n = 357, mean age 72.5 years [SD 7.1]) were available at baseline. Adjusting for age, sex, education and vascular risk factors, there were significant diabetes × time interactions for verbal memory (β -0.06; 95% CI -0.09, -0.02) and verbal fluency (β -0.03; 95% CI -0.06, -0.00). Although people with diabetes had lower brain (β -14.273; 95% CI -21.197, -6.580) and greater ventricular (β 2.672; 95% CI 0.152, 5.193) volumes at baseline, there were no significant diabetes × time interactions (p > 0.05) or evidence of mediation of the diabetes-cognition relationship by brain atrophy. CONCLUSIONS/INTERPRETATION In older community-dwelling people, type 2 diabetes is associated with decline in verbal memory and fluency over ~5 years. The effect of diabetes on brain atrophy may begin earlier (midlife).
Collapse
Affiliation(s)
- Michele L Callisaya
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Private Bag 23, Hobart, TAS, 7000, Australia.
- Peninsula Clinical School, Central Clinical School, Monash University, Melbourne, VIC, Australia.
- Stroke and Aging Research Group, Department of Medicine, School of Clinical Sciences, Monash University, Melbourne, VIC, Australia.
| | - Richard Beare
- Peninsula Clinical School, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Developmental Imaging, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Chris Moran
- Peninsula Clinical School, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Geriatric Medicine, Frankston Hospital, Peninsula Health, Melbourne, VIC, Australia
- Department of Aged Care, Alfred Health, Melbourne, VIC, Australia
| | - Thanh Phan
- Stroke and Aging Research Group, Department of Medicine, School of Clinical Sciences, Monash University, Melbourne, VIC, Australia
| | - Wei Wang
- Peninsula Clinical School, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Velandai K Srikanth
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Private Bag 23, Hobart, TAS, 7000, Australia
- Peninsula Clinical School, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Geriatric Medicine, Frankston Hospital, Peninsula Health, Melbourne, VIC, Australia
| |
Collapse
|
93
|
Shinohara M, Sato N. The Roles of Apolipoprotein E, Lipids, and Glucose in the Pathogenesis of Alzheimer’s Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1128:85-101. [DOI: 10.1007/978-981-13-3540-2_5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
94
|
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
|
95
|
Raman MR, Himali JJ, Conner SC, DeCarli C, Vasan RS, Beiser AS, Seshadri S, Maillard P, Satizabal CL. Circulating Vascular Growth Factors and Magnetic Resonance Imaging Markers of Small Vessel Disease and Atrophy in Middle-Aged Adults. Stroke 2018; 49:2227-2229. [PMID: 30354979 PMCID: PMC6101979 DOI: 10.1161/strokeaha.118.022613] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 07/17/2018] [Indexed: 11/16/2022]
Abstract
Background and Purpose- Little is known about associations between vascular growth factors and magnetic resonance imaging (MRI) markers in midlife. We investigated the association of serum VEGF (vascular endothelial growth factor), Ang2 (angiopoietin 2), sTie2 (soluble tyrosine kinase with immunoglobulin-like and EGF-like domains 2), and HGF (hepatocyte growth factor) concentrations with MRI markers of brain aging in middle-aged adults. Methods- We evaluated 1853 participants (mean age, 46±9 years; 46% men) from the Framingham Heart Study. Serum growth factor concentrations were measured using standardized immunoassays. Outcomes included total brain, cortical and subcortical gray matter, white matter, cerebrospinal fluid, and white matter hyperintensity volumes derived from MRI; as well as fractional anisotropy in white matter tracts from diffusion tensor imaging. We related VEGF, Ang2, sTie2, and HGF to MRI measures using multivariable regression models adjusting for vascular risk factors. We tested for interactions with APOE (apolipoprotein E) genotype and CRP (C-reactive protein). Results were corrected for multiple comparisons. Results- Higher sTie2 was associated with smaller total brain (estimate by SD unit±SE=-0.08±0.02, P=0.002) and larger white matter hyperintensity (0.08±0.02, P=0.002) volumes. Furthermore, higher Ang2 (0.06±0.02, P=0.049) and HGF (0.09±0.02, P=0.001) were associated with larger cerebrospinal fluid volumes. Finally, higher Ang2 was associated with decreased fractional anisotropy, in APOE-ε4 carriers only. Conclusions- Vascular growth factors are associated with early MRI markers of small vessel disease and neurodegeneration in middle-aged adults.
Collapse
Affiliation(s)
- Mekala R Raman
- From the Department of Neurology (M.R.R., J.J.H., A.S.B., S.S., C.L.S.)
- Boston University School of Medicine, MA; Framingham Heart Study, MA (M.R.R., J.J.H., S.C.C., R.S.V., A.S.B., S.S., C.L.S.)
| | - Jayandra J Himali
- From the Department of Neurology (M.R.R., J.J.H., A.S.B., S.S., C.L.S.)
- Boston University School of Medicine, MA; Framingham Heart Study, MA (M.R.R., J.J.H., S.C.C., R.S.V., A.S.B., S.S., C.L.S.)
- Department of Biostatistics (J.J.H., S.C.C., A.S.B.)
| | - Sarah C Conner
- Boston University School of Medicine, MA; Framingham Heart Study, MA (M.R.R., J.J.H., S.C.C., R.S.V., A.S.B., S.S., C.L.S.)
- Department of Biostatistics (J.J.H., S.C.C., A.S.B.)
| | - Charles DeCarli
- Boston University School of Public Health, MA; Department of Neurology, UC Davis School of Medicine, Sacramento, CA (C.D., P.M.)
| | - Ramachandran S Vasan
- Department of Medicine (R.S.V.)
- Boston University School of Medicine, MA; Framingham Heart Study, MA (M.R.R., J.J.H., S.C.C., R.S.V., A.S.B., S.S., C.L.S.)
- Department of Epidemiology (R.S.V.)
| | - Alexa S Beiser
- From the Department of Neurology (M.R.R., J.J.H., A.S.B., S.S., C.L.S.)
- Boston University School of Medicine, MA; Framingham Heart Study, MA (M.R.R., J.J.H., S.C.C., R.S.V., A.S.B., S.S., C.L.S.)
- Department of Biostatistics (J.J.H., S.C.C., A.S.B.)
| | - Sudha Seshadri
- From the Department of Neurology (M.R.R., J.J.H., A.S.B., S.S., C.L.S.)
- Boston University School of Medicine, MA; Framingham Heart Study, MA (M.R.R., J.J.H., S.C.C., R.S.V., A.S.B., S.S., C.L.S.)
- Department of Neurology (S.S.)
- Department of Psychiatry (S.S.)
- Department of Cellular and Integrative Physiology (S.S.)
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, TX (S.S., C.L.S.)
| | - Pauline Maillard
- Boston University School of Public Health, MA; Department of Neurology, UC Davis School of Medicine, Sacramento, CA (C.D., P.M.)
| | - Claudia L Satizabal
- From the Department of Neurology (M.R.R., J.J.H., A.S.B., S.S., C.L.S.)
- Boston University School of Medicine, MA; Framingham Heart Study, MA (M.R.R., J.J.H., S.C.C., R.S.V., A.S.B., S.S., C.L.S.)
- Department of Epidemiology & Biostatistics (C.L.S.)
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, TX (S.S., C.L.S.)
| |
Collapse
|
96
|
Wu M, Fatukasi O, Yang S, Alger J, Barker PB, Hetherington H, Kim T, Levine A, Martin E, Munro CA, Parrish T, Ragin A, Sacktor N, Seaberg E, Becker JT. HIV disease and diabetes interact to affect brain white matter hyperintensities and cognition. AIDS 2018; 32:1803-1810. [PMID: 29794829 PMCID: PMC6082131 DOI: 10.1097/qad.0000000000001891] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Since the onset of combination antiretroviral therapy use, the incidence of HIV-associated dementia and of HIV encephalitis has fallen dramatically. The present study investigates the extent of white matter hyperintensities (WMHs) among individuals with HIV disease, and factors that predict their presence and their impact on psychomotor speed. METHODS A total of 322 men participating in the Multicenter AIDS Cohort Study (185 HIV-infected, age: 57.5 ± 6.0) underwent MRI scans of the brain. T1-weighted magnetization-prepared rapid gradient-echo (MP-RAGE) and T2-weighted Fluid Attenuated Inversion Recovery (FLAIR) images were obtained and processed using an automated method for identifying and measuring WMHs. WMH burden was expressed as the log10 transformed percentage of total white matter. RESULTS There were no significant associations between WMHs and HIV disease. However, the extent of WMHs was predicted by age more than 60 (β = 0.17), non-white race (β = 0.14), glomerular filtration rate (β = -0.11), and the presence of diabetes (β = 0.12). There were no interactions between HIV status and age (β = -0.03) or between age and diabetes (β = 0.07). However, the interaction between HIV infection and diabetes was significant (β = 0.26). The extent of WMHs was significantly associated with performance on measures of psychomotor speed (β = 0.15). CONCLUSION In today's therapeutic environment, in HIV-infected and HIV seronegative individuals, those factors which affect the cerebrovasculature are the best predictors of WMHs. Diabetes has a specific impact among HIV-infected, but not uninfected, men, suggesting the need for more aggressive treatment even in the prediabetes state, especially as WMHs affect cognitive functions.
Collapse
Affiliation(s)
- Minjie Wu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Omalara Fatukasi
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Shaolin Yang
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois
| | - Jeffery Alger
- Department of Radiology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California
| | - Peter B Barker
- Departments of Radiology, The Johns Hopkins University, Baltimore, Maryland
| | - Hoby Hetherington
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Tae Kim
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Andrew Levine
- Department of Neurology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California
| | - Eileen Martin
- Department of Psychiatry, Rush University Medical School, Chicago, Illinois
| | - Cynthia A Munro
- Department of Neurology, The Johns Hopkins University, Baltimore, Maryland
| | - Todd Parrish
- Department of Radiology, Northwestern University, Evanston, Illinois
| | - Ann Ragin
- Department of Radiology, Northwestern University, Evanston, Illinois
| | - Ned Sacktor
- Department of Neurology, The Johns Hopkins University, Baltimore, Maryland
| | - Eric Seaberg
- Department of Epidemiology, The Johns Hopkins University, Baltimore, Maryland
| | - James T Becker
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurology
- Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
97
|
Prediabetes and diabetes accelerate cognitive decline and predict microvascular lesions: A population-based cohort study. Alzheimers Dement 2018; 15:25-33. [DOI: 10.1016/j.jalz.2018.06.3060] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 06/13/2018] [Accepted: 06/27/2018] [Indexed: 02/06/2023]
|
98
|
Holingue C, Wennberg A, Berger S, Polotsky VY, Spira AP. Disturbed sleep and diabetes: A potential nexus of dementia risk. Metabolism 2018; 84:85-93. [PMID: 29409842 PMCID: PMC5995651 DOI: 10.1016/j.metabol.2018.01.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 01/19/2018] [Accepted: 01/26/2018] [Indexed: 12/28/2022]
Abstract
Type 2 diabetes (T2D) and sleep disturbance (e.g., insomnia, sleep-disordered breathing) are prevalent conditions among older adults that are associated with cognitive decline and dementia, including Alzheimer's disease (AD). Importantly, disturbed sleep is associated with alterations in insulin sensitivity and glucose metabolism, and may increase the risk of T2D, and T2D-related complications (e.g., pain, nocturia) can negatively affect sleep. Despite these associations, little is known about how interactions between T2D and sleep disturbance might alter cognitive trajectories or the pathological changes that underlie dementia. Here, we review links among T2D, sleep disturbance, cognitive decline and dementia-including preclinical and clinical AD-and identify gaps in the literature, that if addressed, could have significant implications for the prevention of poor cognitive outcomes.
Collapse
Affiliation(s)
- Calliope Holingue
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, United States.
| | - Alexandra Wennberg
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States.
| | - Slava Berger
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, United States.
| | - Vsevolod Y Polotsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, United States.
| | - Adam P Spira
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, United States; Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, United States; Johns Hopkins Center on Aging and Health, United States.
| |
Collapse
|
99
|
Chatterjee S, Mudher A. Alzheimer's Disease and Type 2 Diabetes: A Critical Assessment of the Shared Pathological Traits. Front Neurosci 2018; 12:383. [PMID: 29950970 PMCID: PMC6008657 DOI: 10.3389/fnins.2018.00383] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/22/2018] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) and Type 2 Diabetes Mellitus (T2DM) are two of the most prevalent diseases in the elderly population worldwide. A growing body of epidemiological studies suggest that people with T2DM are at a higher risk of developing AD. Likewise, AD brains are less capable of glucose uptake from the surroundings resembling a condition of brain insulin resistance. Pathologically AD is characterized by extracellular plaques of Aβ and intracellular neurofibrillary tangles of hyperphosphorylated tau. T2DM, on the other hand is a metabolic disorder characterized by hyperglycemia and insulin resistance. In this review we have discussed how Insulin resistance in T2DM directly exacerbates Aβ and tau pathologies and elucidated the pathophysiological traits of synaptic dysfunction, inflammation, and autophagic impairments that are common to both diseases and indirectly impact Aβ and tau functions in the neurons. Elucidation of the underlying pathways that connect these two diseases will be immensely valuable for designing novel drug targets for Alzheimer's disease.
Collapse
Affiliation(s)
- Shreyasi Chatterjee
- Centre of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Amritpal Mudher
- Centre of Biological Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
100
|
Sun Q, Chen GQ, Wang XB, Yu Y, Hu YC, Yan LF, Zhang X, Yang Y, Zhang J, Liu B, Wang CC, Ma Y, Wang W, Han Y, Cui GB. Alterations of White Matter Integrity and Hippocampal Functional Connectivity in Type 2 Diabetes Without Mild Cognitive Impairment. Front Neuroanat 2018; 12:21. [PMID: 29615873 PMCID: PMC5869188 DOI: 10.3389/fnana.2018.00021] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 03/06/2018] [Indexed: 12/26/2022] Open
Abstract
Aims: To investigate the white matter (WM) integrity and hippocampal functional connectivity (FC) in type 2 diabetes mellitus (T2DM) patients without mild cognitive impairment (MCI) by using diffusion tensor imaging (DTI) and resting-state functional magnetic resonance imaging (rs-fMRI), respectively. Methods: Twelve T2DM patients without MCI and 24 age, sex and education matched healthy controls (HC) were recruited. DTI and rs-fMRI data were subsequently acquired on a 3.0T MR scanner. Tract-based spatial statistics (TBSS) combining region of interests (ROIs) analysis was used to investigate the alterations of DTI metrics (fractional anisotropy (FA), mean diffusivity (MD), λ1 and λ23) and FC measurement was performed to calculate hippocampal FC with other brain regions. Cognitive function was evaluated by using Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment (MoCA). Brain volumes were also evaluated among these participants. Results: There were no difference of MMSE and MoCA scores between two groups. Neither whole brain nor regional brain volume decrease was revealed in T2DM patients without MCI. DTI analysis revealed extensive WM disruptions, especially in the body of corpus callosum (CC). Significant decreases of hippocampal FC with certain brain structures were revealed, especially with the bilateral frontal cortex. Furthermore, the decreased FA in left posterior thalamic radiation (PTR) and increased MD in the splenium of CC were closely related with the decreased hippocampal FC to caudate nucleus and frontal cortex. Conclusions: T2DM patients without MCI showed extensive WM disruptions and abnormal hippocampal FC. Moreover, the WM disruptions and abnormal hippocampal FC were closely associated. HighlightsT2DM patients without MCI demonstrated no obvious brain volume decrease. Extensive white matter disruptions, especially within the body of corpus callosum, were revealed with DTI analysis among the T2DM patients. Despite no MCI in T2DM patients, decreased functional connectivity between hippocampal region and some critical brain regions were detected. The alterations in hippocampal functional connectivity were closely associated with those of the white matter structures in T2DM patients.
This trial was registered to ClinicalTrials.gov (NCT02420470, https://www.clinicaltrials.gov/).
Collapse
Affiliation(s)
- Qian Sun
- Department of Radiology & Functional and Molecular Imaging, Key Lab of Shaanxi Province, Tangdu Hospital, The Military Medical University of PLA Airforce (Fourth Military Medical University), Xi'an, China
| | - Guan-Qun Chen
- Department of Neurology, XuanWu Hospital, Capital Medical University, Beijing, China
| | - Xi-Bin Wang
- Department of Medical Image Diagnosis, Hanzhong Central Hospital, Hanzhong, China
| | - Ying Yu
- Department of Radiology & Functional and Molecular Imaging, Key Lab of Shaanxi Province, Tangdu Hospital, The Military Medical University of PLA Airforce (Fourth Military Medical University), Xi'an, China
| | - Yu-Chuan Hu
- Department of Radiology & Functional and Molecular Imaging, Key Lab of Shaanxi Province, Tangdu Hospital, The Military Medical University of PLA Airforce (Fourth Military Medical University), Xi'an, China
| | - Lin-Feng Yan
- Department of Radiology & Functional and Molecular Imaging, Key Lab of Shaanxi Province, Tangdu Hospital, The Military Medical University of PLA Airforce (Fourth Military Medical University), Xi'an, China
| | - Xin Zhang
- Department of Radiology & Functional and Molecular Imaging, Key Lab of Shaanxi Province, Tangdu Hospital, The Military Medical University of PLA Airforce (Fourth Military Medical University), Xi'an, China
| | - Yang Yang
- Department of Radiology & Functional and Molecular Imaging, Key Lab of Shaanxi Province, Tangdu Hospital, The Military Medical University of PLA Airforce (Fourth Military Medical University), Xi'an, China
| | - Jin Zhang
- Department of Radiology & Functional and Molecular Imaging, Key Lab of Shaanxi Province, Tangdu Hospital, The Military Medical University of PLA Airforce (Fourth Military Medical University), Xi'an, China
| | - Bin Liu
- Student Brigade, The Military Medical University of PLA Airforce (Fourth Military Medical University), Xi'an, China
| | - Cong-Cong Wang
- Student Brigade, The Military Medical University of PLA Airforce (Fourth Military Medical University), Xi'an, China
| | - Yi Ma
- Student Brigade, The Military Medical University of PLA Airforce (Fourth Military Medical University), Xi'an, China
| | - Wen Wang
- Department of Radiology & Functional and Molecular Imaging, Key Lab of Shaanxi Province, Tangdu Hospital, The Military Medical University of PLA Airforce (Fourth Military Medical University), Xi'an, China
| | - Ying Han
- Department of Neurology, XuanWu Hospital, Capital Medical University, Beijing, China
| | - Guang-Bin Cui
- Department of Radiology & Functional and Molecular Imaging, Key Lab of Shaanxi Province, Tangdu Hospital, The Military Medical University of PLA Airforce (Fourth Military Medical University), Xi'an, China
| |
Collapse
|