101
|
Wang D, Liu L, Li S, Wang C. Effects of paeoniflorin on neurobehavior, oxidative stress, brain insulin signaling, and synaptic alterations in intracerebroventricular streptozotocin-induced cognitive impairment in mice. Physiol Behav 2018; 191:12-20. [PMID: 29572012 DOI: 10.1016/j.physbeh.2018.03.016] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 02/09/2018] [Accepted: 03/14/2018] [Indexed: 01/13/2023]
Abstract
Paeoniflorin (PF) is a natural monoterpene glycoside in Paeonia lactiflora pall with anti-diabetic, antioxidant, anti-inflammatory, and neuro-protective properties. This study was designed to investigate the neuroprotective effects of PF against cognitive deficits induced by intracerebroventricular (ICV) injection of streptozotocin (STZ) in mice. STZ was injected twice intracerebroventrically (3 mg/kg ICV) on alternate days (day 1 and day 3) in mice. Daily treatment with PF (10 mg/kg per day, intraperitoneally) starting from the first dose of STZ for 21 days showed an improvement in ICV-STZ induced cognitive deficits as assessed by novel object recognition and Morris water maze (MWM) test. PF significantly attenuated STZ induced mitochondrial dysfunction manifested by dramatically elevated cytochrome c oxidase activity and ATP synthesis, and restoration of the mitochondrial membrane potential (MMP), and oxidative stress in hippocampus and in the cortex compared to control. Moreover, PF treatment also markedly increased synaptic density in the CA1 region of the hippocampus compared to control. Furthermore, PF ameliorated defective insulin signaling by up-regulating p-PI3K and p-Akt protein expression while downregulating p-IRS-1 protein expression. Taken together, the outcomes of the current study suggest the therapeutic potential of PF in the cognitive deficits induced by ICV-STZ.
Collapse
Affiliation(s)
- Dongmei Wang
- Department of Pathogen Biology, Medical College, Henan University of Science and Technology, Luoyang, China.
| | - Ling Liu
- Department of Pharmacy, Medical College, Henan University of Science and Technology, Luoyang, China
| | - Sanqiang Li
- Department of Biochemistry and Molecular Biology, Medical College, Henan University of Science and Technology, Luoyang, China.
| | - Chenying Wang
- Department of Pathogen Biology, Medical College, Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
102
|
Ekblad LL, Johansson J, Helin S, Viitanen M, Laine H, Puukka P, Jula A, Rinne JO. Midlife insulin resistance, APOE genotype, and late-life brain amyloid accumulation. Neurology 2018; 90:e1150-e1157. [PMID: 29476033 PMCID: PMC5880630 DOI: 10.1212/wnl.0000000000005214] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 12/21/2017] [Indexed: 01/01/2023] Open
Abstract
Objective To examine whether midlife insulin resistance is an independent risk factor for brain amyloid accumulation in vivo after 15 years, and whether this risk is modulated by APOE ε4 genotype. Methods This observational study examined 60 elderly volunteers without dementia (mean age at baseline 55.4 and at follow-up 70.9 years, 55.5% women) from the Finnish population-based, nationwide Health2000 study with [11C]Pittsburgh compound B–PET imaging in 2014–2016. The participants were recruited according to their homeostatic model assessment of insulin resistance (HOMA-IR) values in the year 2000, and their APOE ε4 genotype. The exposure group (IR+, n = 30) consisted of individuals with HOMA-IR >2.17 at baseline (highest tertile of the Health2000 study population), and the control group (IR−, n = 30) consisted of individuals with HOMA-IR <1.25 at baseline (lowest tertile). The groups were enriched for APOE ε4 carriers, resulting in 50% (n = 15) APOE ε4 carriers in both groups. Analyses were performed with multivariate logistic and linear regression. Results An amyloid-positive PET scan was found in 33.3% of the IR− group and 60.0% of the IR+ group (odds ratio 3.0, 95% confidence interval 1.1–8.9, p = 0.04). The increased risk was seen in carriers and noncarriers of APOE ε4 genotype. Higher midlife, but not late-life continuous HOMA-IR was associated with a greater brain amyloid burden at follow-up after multivariate adjustments for other cognitive and metabolic risk factors (β = 0.11, 95% confidence interval 0.002–0.22, p = 0.04). Conclusions These results indicate that midlife insulin resistance is an independent risk factor for brain amyloid accumulation in elderly individuals without dementia.
Collapse
Affiliation(s)
- Laura L Ekblad
- From the Turku PET Centre (L.L.E., J.J., S.H., J.O.R.) and Department of Geriatrics (M.V., H.L.), Turku City Hospital (M.V., H.L.), University of Turku, Finland; Department of Radiation Sciences (J.J.), Umeå University; Clinical Geriatrics (M.V.), Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden; Department of Medicine, University of Turku (H.L.), and Division of Clinical Neurosciences (J.O.R.), Turku University Hospital; and National Institute for Health and Welfare (P.P., A.J.), Turku, Finland.
| | - Jarkko Johansson
- From the Turku PET Centre (L.L.E., J.J., S.H., J.O.R.) and Department of Geriatrics (M.V., H.L.), Turku City Hospital (M.V., H.L.), University of Turku, Finland; Department of Radiation Sciences (J.J.), Umeå University; Clinical Geriatrics (M.V.), Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden; Department of Medicine, University of Turku (H.L.), and Division of Clinical Neurosciences (J.O.R.), Turku University Hospital; and National Institute for Health and Welfare (P.P., A.J.), Turku, Finland
| | - Semi Helin
- From the Turku PET Centre (L.L.E., J.J., S.H., J.O.R.) and Department of Geriatrics (M.V., H.L.), Turku City Hospital (M.V., H.L.), University of Turku, Finland; Department of Radiation Sciences (J.J.), Umeå University; Clinical Geriatrics (M.V.), Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden; Department of Medicine, University of Turku (H.L.), and Division of Clinical Neurosciences (J.O.R.), Turku University Hospital; and National Institute for Health and Welfare (P.P., A.J.), Turku, Finland
| | - Matti Viitanen
- From the Turku PET Centre (L.L.E., J.J., S.H., J.O.R.) and Department of Geriatrics (M.V., H.L.), Turku City Hospital (M.V., H.L.), University of Turku, Finland; Department of Radiation Sciences (J.J.), Umeå University; Clinical Geriatrics (M.V.), Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden; Department of Medicine, University of Turku (H.L.), and Division of Clinical Neurosciences (J.O.R.), Turku University Hospital; and National Institute for Health and Welfare (P.P., A.J.), Turku, Finland
| | - Hanna Laine
- From the Turku PET Centre (L.L.E., J.J., S.H., J.O.R.) and Department of Geriatrics (M.V., H.L.), Turku City Hospital (M.V., H.L.), University of Turku, Finland; Department of Radiation Sciences (J.J.), Umeå University; Clinical Geriatrics (M.V.), Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden; Department of Medicine, University of Turku (H.L.), and Division of Clinical Neurosciences (J.O.R.), Turku University Hospital; and National Institute for Health and Welfare (P.P., A.J.), Turku, Finland
| | - Pauli Puukka
- From the Turku PET Centre (L.L.E., J.J., S.H., J.O.R.) and Department of Geriatrics (M.V., H.L.), Turku City Hospital (M.V., H.L.), University of Turku, Finland; Department of Radiation Sciences (J.J.), Umeå University; Clinical Geriatrics (M.V.), Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden; Department of Medicine, University of Turku (H.L.), and Division of Clinical Neurosciences (J.O.R.), Turku University Hospital; and National Institute for Health and Welfare (P.P., A.J.), Turku, Finland
| | - Antti Jula
- From the Turku PET Centre (L.L.E., J.J., S.H., J.O.R.) and Department of Geriatrics (M.V., H.L.), Turku City Hospital (M.V., H.L.), University of Turku, Finland; Department of Radiation Sciences (J.J.), Umeå University; Clinical Geriatrics (M.V.), Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden; Department of Medicine, University of Turku (H.L.), and Division of Clinical Neurosciences (J.O.R.), Turku University Hospital; and National Institute for Health and Welfare (P.P., A.J.), Turku, Finland
| | - Juha O Rinne
- From the Turku PET Centre (L.L.E., J.J., S.H., J.O.R.) and Department of Geriatrics (M.V., H.L.), Turku City Hospital (M.V., H.L.), University of Turku, Finland; Department of Radiation Sciences (J.J.), Umeå University; Clinical Geriatrics (M.V.), Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden; Department of Medicine, University of Turku (H.L.), and Division of Clinical Neurosciences (J.O.R.), Turku University Hospital; and National Institute for Health and Welfare (P.P., A.J.), Turku, Finland
| |
Collapse
|
103
|
Shen Y, Xia Y, Meng S, Lim NKH, Wang W, Huang F. SH2B1 is Involved in the Accumulation of Amyloid-β42 in Alzheimer's Disease. J Alzheimers Dis 2018; 55:835-847. [PMID: 27802221 DOI: 10.3233/jad-160233] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is characterized by deficits in learning and memory abilities, as well as pathological changes of amyloid-β (Aβ) plaque and neurofibrillary tangle formation in the brain. Insulin has been identified as a modulator of the neuronal pathways involved in learning and memory, and is also implicated as a modulator of Aβ and tau metabolism. Disrupted insulin signaling pathways are evident in AD patients and it is understood that type 2 diabetes can increase the risk of developing AD, suggesting a possible link between metabolic disorders and neurodegeneration. SH2B1 is a key protein in the insulin signaling pathway involved in regulating the activity of the insulin receptor. To further identify the role of the insulin signaling pathway in the pathology of AD, SH2B (dSH2B homologue in flies) in neurons was partially knocked out or overexpressed in an AD Drosophila model expressing Aβ42. Partial knockout of neuronal SH2B in the Aβ42-expressing Drosophila had a detrimental effect on mobility and neurotransmission, and increased levels and intraneuronal accumulation of Aβ42, as assessed by ELISA and immunostaining. Alternatively, partial overexpression of neuronal SH2B in the Aβ42-expressing Drosophila improved lifespan, mobility, and neurotransmission, as well as decreased levels and intraneuronal accumulation of Aβ42. Thus, SH2B1 may be an upstream modulator of Aβ metabolism, acting to inhibit Aβ accumulation, and has a role in the pathogenesis of AD. SH2B1 may therefore have potential as a therapeutic target for this common form of dementia.
Collapse
Affiliation(s)
- Yijun Shen
- Department of Neurology, Xin Hua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Advanced Research Institute, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yiling Xia
- Department of Neurology, Xin Hua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Advanced Research Institute, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shiquan Meng
- Shanghai Advanced Research Institute, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Nastasia K H Lim
- Shanghai Advanced Research Institute, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Wenan Wang
- Department of Neurology, Xin Hua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China.,Department of Neurology, Xin Hua Hospital Chongming Branch Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fude Huang
- Shanghai Advanced Research Institute, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
104
|
Martins IV, Rivers-Auty J, Allan SM, Lawrence CB. Mitochondrial Abnormalities and Synaptic Loss Underlie Memory Deficits Seen in Mouse Models of Obesity and Alzheimer's Disease. J Alzheimers Dis 2018; 55:915-932. [PMID: 27802235 PMCID: PMC5278950 DOI: 10.3233/jad-160640] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Obesity is associated with impaired memory in humans, and obesity induced by high-fat diets leads to cognitive deficits in rodents and in mouse models of Alzheimer's disease (AD). However, it remains unclear how high-fat diets contribute to memory impairment. Therefore, we tested the effect of a high-fat diet on memory in male and female control non-transgenic (Non-Tg) and triple-transgenic AD (3xTgAD) mice and determined if a high-fat diet caused similar ultrastructural abnormalities to those observed in AD. Behavior was assessed in mice on control or high-fat diet at 4, 8, or 14 months of age and ultrastructural analysis at 8 months of age. A high-fat diet increased body weight, fat weight, and insulin levels with some differences in these metabolic responses observed between Non-Tg and 3xTgAD mice. In both sexes, high-fat feeding caused memory impairments in Non-Tg mice and accelerated memory deficits in 3xTgAD mice. In 3xTgAD mice, changes in hippocampal mitochondrial morphology were observed in capillaries and brain neuropil that were accompanied by a reduction in synapse number. A high-fat diet also caused mitochondria abnormalities and a reduction in synapse number in Non-Tg mice, but did not exacerbate the changes seen in 3xTgAD mice. Our data demonstrate that a high-fat diet affected memory in Non-Tg mice and produced similar impairments in mitochondrial morphology and synapse number comparable to those seen in AD mice, suggesting that the detrimental effects of a high-fat diet on memory might be due to changes in mitochondrial morphology leading to a reduction in synaptic number.
Collapse
Affiliation(s)
| | | | | | - Catherine B. Lawrence
- Correspondence to: Catherine B. Lawrence, PhD, Faculty of Life Sciences, A.V. Hill Building, University of Manchester, Oxford Road, Manchester, M13 9PT, UK. Tel.: +44 161 275 5253; E-mail:
| |
Collapse
|
105
|
Ding B, Xiao R, Ma W, Zhao L, Bi Y, Zhang Y. The association between macronutrient intake and cognition in individuals aged under 65 in China: a cross-sectional study. BMJ Open 2018; 8:e018573. [PMID: 29317416 PMCID: PMC5781185 DOI: 10.1136/bmjopen-2017-018573] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE The aim of this retrospective study was to explore the correlation between daily energy intake from macronutrients and cognitive functions in a Chinese population aged less than 65 years. DESIGN This is a cross-sectional study to explore the relationships between macronutrients' intake and cognitive function. The analysis of variance (ANOVA) and χ2 test were used to compare the demographic and physical characteristics, lifestyle and laboratory parameters with the intake of macronutrients among different quartiles of % fat/energy. Multivariate logistic regression analysis was applied to identify the potential risk factors of mild cognitive impairment (MCI). PARTICIPANTS Young and middle-aged participants (age <65 years) were recruited from Beijing, China. The Montreal cognitive assessment (MoCA) and mini-mental state examination (MMSE) were used to evaluate the cognitive functions, and the dietary intake of the participants was estimated with a semi-quantitative food frequency questionnaire (FFQ). RESULTS Among the 661 participants, 80 (12.1%) had MCI, while 581 (87.9%) had normal cognitive functions. On evaluating the data based on the age group, educational background, and conditions of hyperlipidaemia and total energy intake, the results revealed that high % fat (upper quartile: adjusted OR (aOR) 3.90, 95% CI1.53 to 9.89, P=0.004), and high % protein intake (upper quartile: aOR 2.77, 95% CI 1.24 to 6.15) were greatly associated with increased frequency of MCI, while high % carbohydrate intake (upper quartile: aOR0.30, 95% CI 0.12 to 0.72) was correlated with decreased prevalence of MCI. CONCLUSION The dietary pattern with high percentage of energy intake from fat and protein, and low-energy intake from carbohydrate might have been associated with cognitive decline in a Chinese population under 65 years of age.
Collapse
Affiliation(s)
- Bingjie Ding
- Department of Clinical Nutrition, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Rong Xiao
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Weiwei Ma
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Lei Zhao
- Department of Molecular Physiology and Biophysics, Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa, USA
| | - Yanxia Bi
- Department of Clinical Nutrition, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yong Zhang
- Department of Chronic Disease, Daxing Centre for Disease Control and Prevention, Beijing, China
| |
Collapse
|
106
|
Rittschof CC, Vekaria HJ, Palmer JH, Sullivan PG. Brain mitochondrial bioenergetics change with rapid and prolonged shifts in aggression in the honey bee, Apis mellifera. J Exp Biol 2018; 221:jeb.176917. [DOI: 10.1242/jeb.176917] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 02/26/2018] [Indexed: 12/12/2022]
Abstract
Neuronal function demands high-level energy production, and as such, a decline in mitochondrial respiration characterizes brain injury and disease. A growing number of studies, however, link brain mitochondrial function to behavioral modulation in non-diseased contexts. In the honey bee, we show for the first time that an acute social interaction, which invokes an aggressive response, may also cause a rapid decline in brain mitochondrial bioenergetics. The degree and speed of this decline has only been previously observed in the context of brain injury. Furthermore, in the honey bee, age-related increases in aggressive tendency are associated with increased baseline brain mitochondrial respiration, as well as increased plasticity in response to metabolic fuel type in vitro. Similarly, diet restriction and ketone body feeding, which commonly enhance mammalian brain mitochondrial function in vivo, cause increased aggression. Thus, even in normal behavioral contexts, brain mitochondria show a surprising degree of variation in function over both rapid and prolonged timescales, with age predicting both baseline function and plasticity in function. These results suggest that mitochondrial function is integral to modulating aggression-related neuronal signaling. We hypothesize that variation in function reflects mitochondrial calcium buffering activity, and that shifts in mitochondrial function signal to the neuronal soma to regulate gene expression and neural energetic state. Modulating brain energetic state is emerging as a critical component of the regulation of behavior in non-diseased contexts.
Collapse
Affiliation(s)
- Clare C. Rittschof
- Department of Entomology, University of Kentucky, S-225 Ag. Science Center North, Lexington, KY, 40546, USA
| | - Hemendra J. Vekaria
- Spinal Cord and Brain Injury Research Center and the Department of Neuroscience, University of Kentucky, 741 South Limestone Street, 475 BBSRB, Lexington, KY 40536-0509, USA
| | - Joseph H. Palmer
- Department of Entomology, University of Kentucky, S-225 Ag. Science Center North, Lexington, KY, 40546, USA
| | - Patrick G. Sullivan
- Spinal Cord and Brain Injury Research Center and the Department of Neuroscience, University of Kentucky, 741 South Limestone Street, 475 BBSRB, Lexington, KY 40536-0509, USA
| |
Collapse
|
107
|
Cezaretto A, Suemoto CK, Bensenor I, Lotufo PA, de Almeida-Pititto B, Ferreira SRG, the ELSA Research Group AquinoEstela M. L.MotaEduardo L. A.BarretoSandhi MariaPassosValéria Maria AzeredoChorDóraCarvalhoMarilia S.BensenorIsabela M.LotufoPaulo A.MillJosé GeraldoDel Carmen MolinaMariaDuncanBruce B.SchmidtMaria InêsSzkloMoyses. Association of adiponectin with cognitive function precedes overt diabetes in the Brazilian Longitudinal Study of Adult Health: ELSA. Diabetol Metab Syndr 2018; 10:54. [PMID: 30002734 PMCID: PMC6038247 DOI: 10.1186/s13098-018-0354-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 06/28/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Adiponectin is an insulin-sensitizer adipocytokine endowed with neuroprotective actions. Whether adiponectin regulates neuronal functioning toward delaying cognitive decline independently of the glucose metabolism disturbance has been poorly explored. This study evaluated if the performance in cognitive tests was associated with adiponectin levels prior the development of type 2 diabetes in middle-aged individuals. METHODS A sample of 938 non-diabetic participants of ELSA had their cognitive function assessed by the CERAD delayed word recall test, the verbal fluency test and the trail making test. Stepwise multiple linear regression using forward selection had the response to cognitive tests as the dependent variable and adiponectin as the independent variable of main interest, adjusted for glucose tolerance status and confounders. RESULTS Mean age was 45.7 ± 4.9 years, 54.5% were women, 43.0% had high education level, 59.3% weight excess and 70.0% prediabetes. In crude model, only the delayed recall memory was associated with adiponectin levels. In an initial regression model, delayed recall memory remained independently associated with adiponectin levels and prediabetes. After complete adjustments, adiponectin but not prediabetes maintained independently associated with delayed recall memory (β 0.067; 95% CI 0.006-0.234; p = 0.040). On the other hand, learning memory showed to be associated with prediabetes (β 0.71 95% CI 0.17; 1.24; p = 0.009) but not with adiponectin. CONCLUSIONS The association of memory with adiponectin in middle-aged individuals, prior overt diabetes, suggests that this adipocytokine could anticipate cognitive impairmentρ detection, when preventive strategies could be more effectively implemented. The usefulness of adiponectin to identify increased risk for cognitive dysfunction before advanced age needs to be prospectively investigated in ELSA cohort.
Collapse
Affiliation(s)
- Adriana Cezaretto
- Department of Epidemiology, School of Public Health, University of Sao Paulo, São Paulo, Brazil
| | | | - Isabela Bensenor
- Department of Internal Medicine, Medical School, University of Sao Paulo, São Paulo, Brazil
| | - Paulo A. Lotufo
- Department of Internal Medicine, Medical School, University of Sao Paulo, São Paulo, Brazil
| | | | - Sandra R. G. Ferreira
- Department of Epidemiology, School of Public Health, University of Sao Paulo, São Paulo, Brazil
| | | |
Collapse
|
108
|
A maternal high-fat diet during pregnancy and lactation, in addition to a postnatal high-fat diet, leads to metabolic syndrome with spatial learning and memory deficits: beneficial effects of resveratrol. Oncotarget 2017; 8:111998-112013. [PMID: 29340106 PMCID: PMC5762374 DOI: 10.18632/oncotarget.22960] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 11/17/2017] [Indexed: 01/16/2023] Open
Abstract
We tested the hypothesis that high-fat diet consumption during pregnancy, lactation, and/or post weaning, altered the expression of molecular mediators involved in hippocampal synaptic efficacy and impaired spatial learning and memory in adulthood. The beneficial effect of resveratrol was assessed. Dams were fed a rat chow diet or a high-fat diet before mating, during pregnancy, and throughout lactation. Offspring were weaned onto either a rat chow or a high-fat diet. Four experimental groups were generated, namely CC, HC, CH, and HH (maternal chow diet or high-fat diet; postnatal chow diet or high-fat diet). A fifth group fed with HH plus resveratrol (HHR) was generated. Morris water maze test was used to evaluate spatial learning and memory. Blood pressure and IPGTT was measured to assess insulin resistance. Dorsal hippocampal expression of certain biochemical molecules, including sirtuin 1, ERK, PPARγ, adiponectin, and BDNF were measured. Rats in HH group showed impaired spatial memory, which was partly restored by the administration of resveratrol. Rats in HH group also showed impaired glucose tolerance and increased blood pressure, all of which was rescued by resveratrol administration. Additionally, SIRT1, phospho-ERK1/2, and phospho-PPARγ, adiponectin and BDNF were all dysregulated in rats placed in HH group; administration of resveratrol restored the expression and regulation of these molecules. Overall, our results suggest that maternal high-fat diet during pregnancy and/or lactation sensitizes the offspring to the adverse effects of a subsequent high-fat diet on hippocampal function; however, administration of resveratrol is demonstrated to be beneficial in rescuing these effects.
Collapse
|
109
|
Vieira MNN, Lima-Filho RAS, De Felice FG. Connecting Alzheimer's disease to diabetes: Underlying mechanisms and potential therapeutic targets. Neuropharmacology 2017; 136:160-171. [PMID: 29129775 DOI: 10.1016/j.neuropharm.2017.11.014] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/07/2017] [Accepted: 11/08/2017] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) is a risk factor for type 2 diabetes and vice versa, and a growing body of evidence indicates that these diseases are connected both at epidemiological, clinical and molecular levels. Recent studies have begun to reveal common pathogenic mechanisms shared by AD and type 2 diabetes. Impaired neuronal insulin signaling and endoplasmic reticulum (ER) stress are present in animal models of AD, similar to observations in peripheral tissue in T2D. These findings shed light into novel diabetes-related mechanisms leading to brain dysfunction in AD. Here, we review the literature on selected mechanisms shared between these diseases and discuss how the identification of such mechanisms may lead to novel therapeutic targets in AD. This article is part of the Special Issue entitled 'Metabolic Impairment as Risk Factors for Neurodegenerative Disorders.'
Collapse
Affiliation(s)
- Marcelo N N Vieira
- 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.
| | - Ricardo A S Lima-Filho
- Institute of Medical Biochemistry Leopoldo de Meis, 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, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario K7L 3N6, Canada.
| |
Collapse
|
110
|
Neth BJ, Craft S. Insulin Resistance and Alzheimer's Disease: Bioenergetic Linkages. Front Aging Neurosci 2017; 9:345. [PMID: 29163128 PMCID: PMC5671587 DOI: 10.3389/fnagi.2017.00345] [Citation(s) in RCA: 203] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 10/13/2017] [Indexed: 12/14/2022] Open
Abstract
Metabolic dysfunction is a well-established feature of Alzheimer's disease (AD), evidenced by brain glucose hypometabolism that can be observed potentially decades prior to the development of AD symptoms. Furthermore, there is mounting support for an association between metabolic disease and the development of AD and related dementias. Individuals with insulin resistance, type 2 diabetes mellitus (T2D), hyperlipidemia, obesity, or other metabolic disease may have increased risk for the development of AD and similar conditions, such as vascular dementia. This association may in part be due to the systemic mitochondrial dysfunction that is common to these pathologies. Accumulating evidence suggests that mitochondrial dysfunction is a significant feature of AD and may play a fundamental role in its pathogenesis. In fact, aging itself presents a unique challenge due to inherent mitochondrial dysfunction and prevalence of chronic metabolic disease. Despite the progress made in understanding the pathogenesis of AD and in the development of potential therapies, at present we remain without a disease-modifying treatment. In this review, we will discuss insulin resistance as a contributing factor to the pathogenesis of AD, as well as the metabolic and bioenergetic disruptions linking insulin resistance and AD. We will also focus on potential neuroimaging tools for the study of the metabolic dysfunction commonly seen in AD with hopes of developing therapeutic and preventative targets.
Collapse
Affiliation(s)
- Bryan J Neth
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Suzanne Craft
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
111
|
Wennberg AMV, Hagen CE, Gottesman RF, Zipunnikov V, Kaufmann CN, Albert MS, Rebok GW, Kasper JD, Spira AP. Longitudinal association between diabetes and cognitive decline: The National Health and Aging Trends Study. Arch Gerontol Geriatr 2017; 72:39-44. [PMID: 28544945 PMCID: PMC5967846 DOI: 10.1016/j.archger.2017.05.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 03/27/2017] [Accepted: 05/09/2017] [Indexed: 12/29/2022]
Abstract
Type II diabetes mellitus (DM) is associated with increased risk of dementia; however, few studies have examined the longitudinal association between DM and cognitive outcomes in large nationally representative cohorts. We investigated these associations in 7605 participants enrolled in the National Health and Aging Trends Study, a nationally representative prospective study of Medicare beneficiaries ≥65, from 2011 to 2015. Participants or proxy respondents reported DM and dementia diagnosis, and participants completed immediate and delayed recall word list learning tests and the Clock Drawing Test. In multivariable-adjusted generalized linear mixed models, baseline DM diagnosis was associated with decline on immediate and delayed word recall and the Clock Drawing Test. In Cox proportional hazards models, DM also predicted incident dementia in older age groups at baseline. These findings further support the notion that DM is associated with cognitive outcomes, suggesting that treatment and prevention of DM may reduce the risk of these outcomes. However, more studies are needed to better understand whether DM treatments affect this relationship.
Collapse
Affiliation(s)
| | - Clinton E Hagen
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States
| | | | - Vadim Zipunnikov
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, United States
| | | | - Marilyn S Albert
- Department of Neurology, Johns Hopkins School of Medicine, United States
| | - George W Rebok
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, United States; Johns Hopkins Center on Aging and Health, United States
| | - Judith D Kasper
- Johns Hopkins Center on Aging and Health, United States; Department of Health Policy and Management, Johns Hopkins Bloomberg School of Public Health, United States
| | - Adam P Spira
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, United States; Johns Hopkins Center on Aging and Health, United States; Department of Psychiatry and Behavioral Science, Johns Hopkins School of Medicine, United States
| |
Collapse
|
112
|
Guo X, Namekata K, Kimura A, Harada C, Harada T. ASK1 in neurodegeneration. Adv Biol Regul 2017; 66:63-71. [PMID: 28882588 DOI: 10.1016/j.jbior.2017.08.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 08/28/2017] [Accepted: 08/29/2017] [Indexed: 12/11/2022]
Abstract
Neurodegenerative diseases (NDDs) such as glaucoma, multiple sclerosis (MS), Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD) are characterized by the progressive loss of neurons, causing irreversible damage to patients. Longer lifespans may be leading to an increase in the number of people affected by NDDs worldwide. Among the pathways strongly impacting the pathogenesis of NDDs, oxidative stress, a condition that occurs because of an imbalance in oxidant and antioxidant levels, has been known to play a vital role in the pathophysiology of NDDs. One of the molecules activated by oxidative stress is apoptosis signal-regulating kinase 1 (ASK1), which has been shown to play a role in NDDs. ASK1 activation is regulated by multiple steps, including oligomerization, phosphorylation, and protein-protein interactions. In the oxidative stress state, reactive oxygen species (ROS) induce the dissociation of thioredoxin, a protein regulating cellular reduction and oxidation (redox), from the N-terminal region of ASK1, and ASK1 is subsequently activated by the oligomerization and phosphorylation of a critical threonine residue, leading to cell death. Here, we review experimental evidence that links ASK1 signaling with the pathogenesis of several NDDs. We propose that ASK1 may be a new point of therapeutic intervention to prevent or treat NDDs.
Collapse
Affiliation(s)
- Xiaoli Guo
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
| | - Kazuhiko Namekata
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Atsuko Kimura
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Chikako Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takayuki Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
113
|
Tsuneki H, Yoshida H, Endo K, Mori N, Hosoh S, Tsuda M, Wada T, Sasaoka T. Different impacts of acylated and non-acylated long-acting insulin analogs on neural functions in vitro and in vivo. Diabetes Res Clin Pract 2017; 129:62-72. [PMID: 28511140 DOI: 10.1016/j.diabres.2017.03.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/13/2017] [Accepted: 03/28/2017] [Indexed: 01/06/2023]
Abstract
AIMS Centrally administered insulin improves cognitive functions in patients with Alzheimer's disease; however, it remains unknown whether long-acting insulin analogs exert more pronounced effects than insulin. In the present study, we directly compared the effects of insulin and its analogs on neural functions in vitro and in vivo. METHODS Cultured rat cerebral cortical neurons were treated with insulin, insulin glargine U100 (Gla), insulin detemir (Det), or insulin degludec (Deg). Moreover, these drugs were intracerebroventricularly administered to mice. Their efficacies were evaluated by biochemical and behavioral analyses. RESULTS In cultured neurons, insulin, Gla, and Det increased phosphorylation of Akt and enhanced gene expression of brain-derived neurotrophic factor to a similar extent, although Deg was less effective. The effects of Det and Deg, but not insulin and Gla were suppressed by addition of albumin. When the drug was centrally administered, the increasing effects of insulin on the Akt phosphorylation were comparable to those of Gla but greater than those of Det in hippocampus and cerebral cortex of diabetic db/db and non-diabetic db/m+ mice. Moreover, insulin and Gla enhanced memory functions in Y-maze test and suppressed depression-like behavior in forced swim test in normal mice to a similar extent, and these effects were more potent than those of Det. CONCLUSIONS Insulin and Gla have greater impacts on central nervous system than insulin analogs with high albumin sensitivity, such as Det and Deg. These pharmacological profiles should be taken into account for developing an insulin-based therapy to treat Alzheimer's disease.
Collapse
Affiliation(s)
- Hiroshi Tsuneki
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | - Hitomi Yoshida
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Kosuke Endo
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Norihiko Mori
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Shuji Hosoh
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Masaaki Tsuda
- Department of Biological Chemistry, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Tsutomu Wada
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Toshiyasu Sasaoka
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| |
Collapse
|
114
|
Farzampour S, Majdi A, Sadigh-Eteghad S. Intranasal insulin treatment improves memory and learning in a rat amyloid-beta model of Alzheimer's disease. Physiol Int 2017; 103:344-353. [PMID: 28229638 DOI: 10.1556/2060.103.2016.3.7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Recently, insulin has been used as a pro-cognitive agent for the potential treatment of Alzheimer's disease (AD), because of its ability to cross the brain-blood barrier (BBB) by a saturable transport system. This study has been designed to evaluate the effects of intranasal insulin regimen, as a bypass system of BBB, on spatial memory in amyloid-beta (Aβ) model of AD in rat. Unilateral infusion of Aβ25-35 (10 nmol/2 µl/rat) into the lateral ventricular region of brain was used to produce a rat model of AD. After a 24-h recovery period, rats received insulin or vehicle via intraperitoneal or intranasal route (0.1, 0.2, and 0.3 IU) for 14 days. Memory function in rats was assessed by Morris water maze test, with 5 days of training and consequent probe test protocol. Different doses of intraperitoneal insulin did not have a significant effect on learning and memory in AD rats. However, intranasal insulin at doses of 0.2 and 0.3 IU improved the learning and memory in Aβ-received rats. In conclusion, intranasal insulin as a non-invasive strategy improves spatial learning and memory in AD model.
Collapse
Affiliation(s)
- S Farzampour
- 1 Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences , Tabriz, Iran
| | - A Majdi
- 1 Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences , Tabriz, Iran
| | - S Sadigh-Eteghad
- 1 Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences , Tabriz, Iran
| |
Collapse
|
115
|
Hippocampal insulin resistance and altered food decision-making as players on obesity risk. Neurosci Biobehav Rev 2017; 77:165-176. [DOI: 10.1016/j.neubiorev.2017.03.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/17/2017] [Accepted: 03/19/2017] [Indexed: 12/17/2022]
|
116
|
Heylmann ASA, Canever L, Gress K, Gomes ST, Fachim I, Michels C, Stopassoli GC, Mastella GA, Steckert AV, Damiani AP, de Andrade VM, Quevedo J, Zugno AI. Pre-clinical investigation of Diabetes Mellitus as a risk factor for schizophrenia. Behav Brain Res 2017; 326:154-164. [PMID: 28286284 DOI: 10.1016/j.bbr.2017.02.043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 02/21/2017] [Accepted: 02/24/2017] [Indexed: 10/20/2022]
Abstract
This study investigated the behavioral and biochemical parameters of DM1 as a risk factor in an animal model of schizophrenia (SZ). All groups: 1 Control (saline+saline); 2 Alloxan (alloxan+saline); 3 Ketamine (saline+ketamine); 4 (Alloxan+Ketamine) were fasted for a period of 18h before the subsequent induction of DM via a single intraperitoneal (i.p) injection of alloxan (150mg/kg). From the 4th to the 10th days, the animals were injected i.p with ketamine (25mg/kg) or saline, once a day, to induce a model of SZ and 30min after the last administration were subjected to behavioral testing. After, the animals were decapitated and the brain structures were removed. Ketamine induced hyperactivity and in the social interaction, ketamine, alloxan and the association of alloxan+ketamine increased the latency and decreased the number of contacts between animals. The animals from the ketamine, alloxan and alloxan+ketamine groups showed a prepulse startle reflex (PPI) deficit at the three intensities (65, 70 and 75dB). Ketamine was shown to be capable of increasing the activity of acetylcholinesterase (AChE) in the brain structures. Combination of alloxan+ketamine seems to have an exacerbated effect within the cholinergic system. For lipid peroxidation and protein carbonyls, alloxan+ketamine appear to have intensified lipid and protein damage in the three structures. Ketamine and the combination of ketamine+alloxan induced DNA damage in both frequency and damage index. This research found a relationship between DM1 and SZ.
Collapse
Affiliation(s)
- Alexandra S Almeida Heylmann
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000, Criciúma, SC, Brazil
| | - Lara Canever
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000, Criciúma, SC, Brazil
| | - Katia Gress
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000, Criciúma, SC, Brazil
| | - Sarah T Gomes
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000, Criciúma, SC, Brazil
| | - Isadora Fachim
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000, Criciúma, SC, Brazil
| | - Carolina Michels
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000, Criciúma, SC, Brazil
| | - Geórgia C Stopassoli
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000, Criciúma, SC, Brazil
| | - Gustavo A Mastella
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000, Criciúma, SC, Brazil
| | - Amanda V Steckert
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000, Criciúma, SC, Brazil
| | - Adriani P Damiani
- Laboratório de Biologia Celular e Molecular, Programa de Pós-graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Vanessa M de Andrade
- Laboratório de Biologia Celular e Molecular, Programa de Pós-graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - João Quevedo
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000, Criciúma, SC, Brazil; Center for Experimental Models in Psychiatry, Department of Psychiatry and Behavioral Sciences, Medical School, University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Alexandra I Zugno
- Laboratório de Neurociências and Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, 88806-000, Criciúma, SC, Brazil.
| |
Collapse
|
117
|
Geijselaers SLC, Sep SJS, Schram MT, van Boxtel MPJ, Henry RMA, Verhey FRJ, Kroon AA, Schaper NC, Dagnelie PC, van der Kallen CJH, Stehouwer CDA, Biessels GJ. Insulin resistance and cognitive performance in type 2 diabetes - The Maastricht study. J Diabetes Complications 2017; 31:824-830. [PMID: 28319003 DOI: 10.1016/j.jdiacomp.2017.01.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 01/28/2017] [Accepted: 01/31/2017] [Indexed: 10/20/2022]
Abstract
AIMS Type 2 diabetes, hyperinsulinemia, and insulin resistance are associated with cognitive impairment. Experimental studies indicate that insulin signaling in the brain is related to cognitive performance. Here we evaluated whether insulin-related variables contribute to the variance in cognitive performance among individuals with type 2 diabetes. METHODS A total of 806 individuals with type 2 diabetes (mean age 62±8years, HbA1c 6.9±1.1%) completed a neuropsychological test battery. Insulin-related variables evaluated were: fasting plasma insulin, C-peptide, and the Homeostasis Model Assessment (HOMA2-IR; in individuals without insulin treatment; n=641). The unadjusted coefficient of determination (R2), obtained from multiple linear regression analyses, was used to estimate the proportion of variance in cognition explained by insulin-related variables. RESULTS Sex, age, and educational level together explained 18.0% (R2) of the variance in memory function, 26.5% in information processing speed, and 22.8% in executive function and attention. Fasting insulin, C-peptide, or HOMA2-IR did not increase the explained variance (maximum ΔR2 0.3%, P≥0.14). Similar results were obtained when insulin-related variables were added to models that additionally included glycemic control, cardiovascular risk factors, and depression. CONCLUSIONS Our results show that measures of peripheral insulin resistance are unrelated to cognitive performance among individuals with adequately controlled type 2 diabetes.
Collapse
Affiliation(s)
- Stefan L C Geijselaers
- Department of Internal Medicine, Maastricht University Medical Center +, Maastricht, the Netherlands; CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands; Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands.
| | - Simone J S Sep
- Department of Internal Medicine, Maastricht University Medical Center +, Maastricht, the Netherlands; CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Miranda T Schram
- Department of Internal Medicine, Maastricht University Medical Center +, Maastricht, the Netherlands; CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Martin P J van Boxtel
- Department of Psychiatry and Neuropsychology and MHeNS School for Mental Health and Neuroscience, Maastricht University Medical Center +, Maastricht, the Netherlands
| | - Ronald M A Henry
- Department of Internal Medicine, Maastricht University Medical Center +, Maastricht, the Netherlands; CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Frans R J Verhey
- Department of Psychiatry and Neuropsychology and MHeNS School for Mental Health and Neuroscience, Maastricht University Medical Center +, Maastricht, the Netherlands
| | - Abraham A Kroon
- Department of Internal Medicine, Maastricht University Medical Center +, Maastricht, the Netherlands; CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Nicolaas C Schaper
- Department of Internal Medicine, Maastricht University Medical Center +, Maastricht, the Netherlands; CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands; CAPHRI School for Public Health and Primary Care, Maastricht University, Maastricht, the Netherlands
| | - Pieter C Dagnelie
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands; CAPHRI School for Public Health and Primary Care, Maastricht University, Maastricht, the Netherlands; Department of Epidemiology, Maastricht University, Maastricht, the Netherlands
| | - Carla J H van der Kallen
- Department of Internal Medicine, Maastricht University Medical Center +, Maastricht, the Netherlands; CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Coen D A Stehouwer
- Department of Internal Medicine, Maastricht University Medical Center +, Maastricht, the Netherlands; CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Geert Jan Biessels
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
118
|
Ramos VW, Batista LO, Cordeiro EM, Oliveira GV, Albuquerque KT. High-monosaccharide intake inhibits anorexigenic hypothalamic insulin response in male rats. Nutr Neurosci 2017; 21:337-340. [PMID: 28276261 DOI: 10.1080/1028415x.2017.1288339] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE The aim of this research is to evaluate if intake of 20% fructose solution is able to change the anorexigenic hypothalamic insulin action. METHODS Thirty day-old male Wistar rats were randomly assigned to one of the following groups: standard chow and water for the rats (Control group, C) and standard chow and 20% fructose solution for the rats (Fructose group, F).These treatments lasted 8 weeks. Three-month-old rats from group C and F received insulin or saline intracerebroventricular injections for evaluation of 24 h food intake, phosphorylated forms of the IR (p-IR) and Akt (p-Akt) proteins and quantified hypothalamic insulin receptor (IR) and insulin receptor substrate 1 (IRS-1) proteins. RESULTS Insulin injection was able to decrease food intake in group C compared to 0.9% saline. However, insulin infusion failed to inhibit 24 h food intake in group F compared to 0.9% saline. The hypothalamic content of the IRS-1 was 37% higher in group F as well as p-Akt protein was significant higher vs. group C. CONCLUSION We concluded that the 20% fructose solution compromised insulin signaling considering that it inhibited the anorexigenic hypothalamic response to acute injection of this hormone and increase of IRS-1 and p-Akt content.
Collapse
Affiliation(s)
- Viviane Wagner Ramos
- a Programa de Pós-graduação em Nutrição , Universidade Federal do Rio de Janeiro (UFRJ) , Rio de Janeiro , Brasil.,d Macaé Municipal Government , Rio de Janeiro , Brasil
| | - Leandro Oliveira Batista
- a Programa de Pós-graduação em Nutrição , Universidade Federal do Rio de Janeiro (UFRJ) , Rio de Janeiro , Brasil.,c Laboratório de Nutrição Experimental (LABNEX) , Universidade Federal do Rio de Janeiro campus Macaé (UFRJ-Macaé), Macaé, Rio de Janeiro , Brasil
| | - Elisaldo Mendes Cordeiro
- b Programa de Pós-graduação em Produtos Bioativos e Biociências , Universidade Federal do Rio de Janeiro campus Macaé (UFRJ-Macaé), Macaé, Rio de Janeiro , Brasil.,d Macaé Municipal Government , Rio de Janeiro , Brasil
| | - Gustavo Vieira Oliveira
- b Programa de Pós-graduação em Produtos Bioativos e Biociências , Universidade Federal do Rio de Janeiro campus Macaé (UFRJ-Macaé), Macaé, Rio de Janeiro , Brasil
| | - Kelse Tibau Albuquerque
- a Programa de Pós-graduação em Nutrição , Universidade Federal do Rio de Janeiro (UFRJ) , Rio de Janeiro , Brasil.,b Programa de Pós-graduação em Produtos Bioativos e Biociências , Universidade Federal do Rio de Janeiro campus Macaé (UFRJ-Macaé), Macaé, Rio de Janeiro , Brasil.,c Laboratório de Nutrição Experimental (LABNEX) , Universidade Federal do Rio de Janeiro campus Macaé (UFRJ-Macaé), Macaé, Rio de Janeiro , Brasil
| |
Collapse
|
119
|
Luczkowski M. “No screams and cries will convince us that white is white and black is black”, an ode to the defenders of amyloid cascade hypothesis of Alzheimer's disease. Coord Chem Rev 2016. [DOI: 10.1016/j.ccr.2016.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
120
|
Wang YF, Ji XM, Lu GM, Zhang LJ. Resting-state functional MR imaging shed insights into the brain of diabetes. Metab Brain Dis 2016; 31:993-1002. [PMID: 27456459 DOI: 10.1007/s11011-016-9872-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 07/05/2016] [Indexed: 12/21/2022]
Abstract
Diabetes mellitus is a common metabolic disease which is associated with increasing risk for multiple cognitive declines. Alterations in brain functional connectivity are believed to be the mechanisms underlying the cognitive function impairments. During the past decade, resting-state functional magnetic resonance imaging (rs-fMRI) has been developed as a major tool to study brain functional connectivity in vivo. This paper briefly reviews the diabetes-associated cognitive impairment, analysis algorithms and clinical applications of rs-fMRI. We also provide future perspectives of rs-fMRI in diabetes.
Collapse
Affiliation(s)
- Yun Fei Wang
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, No. 305 Zhongshan East Road, Nanjing, Jiangsu Province, 210002, China
| | - Xue Man Ji
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, No. 305 Zhongshan East Road, Nanjing, Jiangsu Province, 210002, China.
| | - Guang Ming Lu
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, No. 305 Zhongshan East Road, Nanjing, Jiangsu Province, 210002, China
| | - Long Jiang Zhang
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, No. 305 Zhongshan East Road, Nanjing, Jiangsu Province, 210002, China.
| |
Collapse
|
121
|
Bahramian A, Rastegar K, Namavar MR, Moosavi M. Insulin potentiates the therapeutic effect of memantine against central STZ-induced spatial learning and memory deficit. Behav Brain Res 2016; 311:247-254. [DOI: 10.1016/j.bbr.2016.05.046] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 05/17/2016] [Accepted: 05/21/2016] [Indexed: 10/21/2022]
|
122
|
An assessment of the central disposition of intranasally administered insulin lispro in the cerebrospinal fluid of healthy volunteers and beagle dogs. Drug Deliv Transl Res 2016; 7:11-15. [DOI: 10.1007/s13346-016-0325-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
123
|
The role of hippocampal insulin signaling on postoperative cognitive dysfunction in an aged rat model of abdominal surgery. Life Sci 2016; 162:87-94. [PMID: 27561842 DOI: 10.1016/j.lfs.2016.08.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 08/11/2016] [Accepted: 08/20/2016] [Indexed: 11/20/2022]
Abstract
AIMS This study aimed to investigate the role of central insulin signaling, including glycogen synthase kinase 3β (GSK-3β), and its therapeutic potential for the prevention of postoperative neurocognitive deficits. MAIN METHODS In non-insulin experiment, aged rats were divided into a sham group and abdominal surgery group. In insulin experiment, sham and surgically treated rats were distributed into two groups: an intranasal denatured insulin-treated group and intranasal insulin-treated group. Insulin administration started the day of surgery and continued for 3days. Fourteen-days after surgery, cognitive function was assessed using a novel object recognition test, followed by measurement of hippocampal levels of pro-inflammatory cytokines, GSK-3β, and phosphorylated GSK-3β (pGSK-3β(ser9)). Under identical conditions, lipopolysaccharide (LPS)-induced cytokine release from isolated hippocampal microglia was also tested. KEY FINDINGS In non-insulin experiment, compared with non-surgical animals, the rats that underwent abdominal surgery showed memory deficits and increased hippocampal cytokine levels. The hippocampal ratio of pGSK-3β(ser9)/GSK-3β decreased after surgery, a ratio that was positively correlated with novel object recognition performance in the testing phase. Insulin experiment revealed that perioperative intranasal insulin administration could restore the surgery-induced hippocampal neuroinflammation and hyperactivation of GSK-3β, and prevent impairment in novel object recognition. Furthermore, ex vivo experiments indicated that intranasal insulin administration, as well as pretreatment with SB216763, a GSK-3β inhibitor, resulted in reduction of the surgery-related microglial hyper-reactivity to LPS. SIGNIFICANCE Our findings in aged rats suggest that surgical procedures could impair central insulin signaling including GSK-3β, which makes the individual more susceptible to hippocampal neuroinflammation and related cognitive disorders.
Collapse
|
124
|
Kullmann S, Heni M, Hallschmid M, Fritsche A, Preissl H, Häring HU. Brain Insulin Resistance at the Crossroads of Metabolic and Cognitive Disorders in Humans. Physiol Rev 2016; 96:1169-209. [PMID: 27489306 DOI: 10.1152/physrev.00032.2015] [Citation(s) in RCA: 393] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ever since the brain was identified as an insulin-sensitive organ, evidence has rapidly accumulated that insulin action in the brain produces multiple behavioral and metabolic effects, influencing eating behavior, peripheral metabolism, and cognition. Disturbances in brain insulin action can be observed in obesity and type 2 diabetes (T2D), as well as in aging and dementia. Decreases in insulin sensitivity of central nervous pathways, i.e., brain insulin resistance, may therefore constitute a joint pathological feature of metabolic and cognitive dysfunctions. Modern neuroimaging methods have provided new means of probing brain insulin action, revealing the influence of insulin on both global and regional brain function. In this review, we highlight recent findings on brain insulin action in humans and its impact on metabolism and cognition. Furthermore, we elaborate on the most prominent factors associated with brain insulin resistance, i.e., obesity, T2D, genes, maternal metabolism, normal aging, inflammation, and dementia, and on their roles regarding causes and consequences of brain insulin resistance. We also describe the beneficial effects of enhanced brain insulin signaling on human eating behavior and cognition and discuss potential applications in the treatment of metabolic and cognitive disorders.
Collapse
Affiliation(s)
- Stephanie Kullmann
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD e.V.), Tübingen, Germany; Department of Internal Medicine IV, University of Tübingen, Tübingen, Germany; Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; and Department of Pharmacy and Biochemistry, Faculty of Science, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Martin Heni
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD e.V.), Tübingen, Germany; Department of Internal Medicine IV, University of Tübingen, Tübingen, Germany; Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; and Department of Pharmacy and Biochemistry, Faculty of Science, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Manfred Hallschmid
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD e.V.), Tübingen, Germany; Department of Internal Medicine IV, University of Tübingen, Tübingen, Germany; Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; and Department of Pharmacy and Biochemistry, Faculty of Science, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD e.V.), Tübingen, Germany; Department of Internal Medicine IV, University of Tübingen, Tübingen, Germany; Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; and Department of Pharmacy and Biochemistry, Faculty of Science, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Hubert Preissl
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD e.V.), Tübingen, Germany; Department of Internal Medicine IV, University of Tübingen, Tübingen, Germany; Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; and Department of Pharmacy and Biochemistry, Faculty of Science, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Hans-Ulrich Häring
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD e.V.), Tübingen, Germany; Department of Internal Medicine IV, University of Tübingen, Tübingen, Germany; Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Tübingen, Germany; and Department of Pharmacy and Biochemistry, Faculty of Science, Eberhard Karls Universität Tübingen, Tübingen, Germany
| |
Collapse
|
125
|
Bertram S, Brixius K, Brinkmann C. Exercise for the diabetic brain: how physical training may help prevent dementia and Alzheimer's disease in T2DM patients. Endocrine 2016; 53:350-63. [PMID: 27160819 DOI: 10.1007/s12020-016-0976-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 04/27/2016] [Indexed: 12/21/2022]
Abstract
Epidemiological studies indicate that patients with type 2 diabetes mellitus (T2DM) are at increased risk of developing dementia/Alzheimer's disease (AD). This review, which is based on recent studies, presents a molecular framework that links the two diseases and explains how physical training could help counteract neurodegeneration in T2DM patients. Inflammatory, oxidative, and metabolic changes in T2DM patients cause cerebrovascular complications and can lead to blood-brain-barrier (BBB) breakdown. Peripherally increased pro-inflammatory molecules can then pass the BBB more easily and activate stress-activated pathways, thereby promoting key pathological features of dementia/AD such as brain insulin resistance, mitochondrial dysfunction, and accumulation of neurotoxic beta-amyloid (Aβ) oligomers, leading to synaptic loss, neuronal dysfunction, and cell death. Ceramides can also pass the BBB, induce pro-inflammatory reactions, and disturb brain insulin signaling. In a vicious circle, oxidative stress and the pro-inflammatory environment intensify, leading to further cognitive decline. Low testosterone levels might be a common risk factor in T2DM and AD. Regular physical exercise reinforces antioxidative capacity, reduces oxidative stress, and has anti-inflammatory effects. It improves endothelial function and might increase brain capillarization. Physical training can further counteract dyslipidemia and reduce increased ceramide levels. It might also improve Aβ clearance by up-regulating Aβ transporters and, in some cases, increase basal testosterone levels. In addition, regular physical activity can induce neurogenesis. Physical training should therefore be emphasized as a part of prevention programs developed for diabetic patients to minimize the risk of the onset of neurodegenerative diseases among this specific patient group.
Collapse
Affiliation(s)
- Sebastian Bertram
- Institute of Cardiovascular Research and Sport Medicine, Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Am Sportpark Müngersdorf 6, 50933, Cologne, Germany
| | - Klara Brixius
- Institute of Cardiovascular Research and Sport Medicine, Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Am Sportpark Müngersdorf 6, 50933, Cologne, Germany
| | - Christian Brinkmann
- Institute of Cardiovascular Research and Sport Medicine, Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Am Sportpark Müngersdorf 6, 50933, Cologne, Germany.
| |
Collapse
|
126
|
P2‐422: Insulin Resistance is Associated With a Decline in Verbal Fluency During 11 Years of Follow‐Up. Alzheimers Dement 2016. [DOI: 10.1016/j.jalz.2016.06.1634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
127
|
Keeney JTR, Ibrahimi S, Zhao L. Human ApoE Isoforms Differentially Modulate Glucose and Amyloid Metabolic Pathways in Female Brain: Evidence of the Mechanism of Neuroprotection by ApoE2 and Implications for Alzheimer's Disease Prevention and Early Intervention. J Alzheimers Dis 2016; 48:411-24. [PMID: 26402005 DOI: 10.3233/jad-150348] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Three major genetic isoforms of apolipoprotein E (ApoE), ApoE2, ApoE3, and ApoE4, exist in humans and lead to differences in susceptibility to Alzheimer's disease (AD). This study investigated the impact of human ApoE isoforms on brain metabolic pathways involved in glucose utilization and amyloid-β (Aβ) degradation, two major areas that are significantly perturbed in preclinical AD. Hippocampal RNA samples from middle-aged female mice with targeted human ApoE2, ApoE3, and ApoE4 gene replacement were comparatively analyzed with a qRT-PCR custom array for the expression of 85 genes involved in insulin/insulin-like growth factor (Igf) signaling. Consistent with its protective role against AD, ApoE2 brain exhibited the most metabolically robust profile among the three ApoE genotypes. When compared to ApoE2 brain, both ApoE3 and ApoE4 brains exhibited markedly reduced levels of Igf1, insulin receptor substrates (Irs), and facilitated glucose transporter 4 (Glut4), indicating reduced glucose uptake. Additionally, ApoE4 brain exhibited significantly decreased Pparg and insulin-degrading enzyme (Ide), indicating further compromised glucose metabolism and Aβ dysregulation associated with ApoE4. Protein analysis showed significantly decreased Igf1, Irs, and Glut4 in ApoE3 brain, and Igf1, Irs, Glut4, Pparg, and Ide in ApoE4 brain compared to ApoE2 brain. These data provide the first documented evidence that human ApoE isoforms differentially affect brain insulin/Igf signaling and downstream glucose and amyloid metabolic pathways, illustrating a potential mechanism for their differential risk in AD. A therapeutic strategy that enhances brain insulin/Igf1 signaling activity to a more robust ApoE2-like phenotype favoring both energy production and amyloid homeostasis holds promise for AD prevention and early intervention.
Collapse
Affiliation(s)
| | - Shaher Ibrahimi
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Liqin Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA.,Neuroscience Graduate Program, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
128
|
Trueba-Saiz A, Torres Aleman I. Insulin-like peptides signaling in Alzheimer's disease: on the road to alternative therapeutics. Curr Opin Behav Sci 2016. [DOI: 10.1016/j.cobeha.2015.11.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
129
|
Noble EE, Kanoski SE. Early life exposure to obesogenic diets and learning and memory dysfunction. Curr Opin Behav Sci 2016; 9:7-14. [PMID: 26858972 DOI: 10.1016/j.cobeha.2015.11.014] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Obesogenic dietary factors, such as simple sugars and saturated fatty acids, have been linked to memory impairments and hippocampal dysfunction. Recent evidence suggests that the brain may be particularly vulnerable to the effects of obesogenic diets during early life periods of rapid growth, maturation, and brain development. Investigations utilizing rodent models indicate that early life exposure to "high fat diets" (40-65% kcal derived from fat) or simple sugars (sucrose or high fructose corn syrup) can impair hippocampus-dependent learning and memory processes. In some cases, these deficits occur independent of obesity and metabolic derangement and can persist into adulthood despite dietary intervention. Various neurobiological mechanisms have been identified that may link early life consumption of obesogenic dietary factors with hippocampal dysfunction, including increased neuroinflammation and reduced neurotrophin mediated regulation of neurogenesis and synaptic plasticity. Age, duration of exposure, and dietary composition are key variables contributing to the interaction between early life diet and cognitive dysfunction, however, more research is needed to unravel the precise critical windows of development and causal dietary factors.
Collapse
Affiliation(s)
- Emily E Noble
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| | - Scott E Kanoski
- Department of Biological Sciences, Human and Evolutionary Biology Section, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
130
|
Diabetes and Alzheimer’s disease crosstalk. Neurosci Biobehav Rev 2016; 64:272-87. [PMID: 26969101 DOI: 10.1016/j.neubiorev.2016.03.005] [Citation(s) in RCA: 175] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 01/26/2016] [Accepted: 03/04/2016] [Indexed: 12/12/2022]
|
131
|
Barone E, Di Domenico F, Cassano T, Arena A, Tramutola A, Lavecchia MA, Coccia R, Butterfield DA, Perluigi M. Impairment of biliverdin reductase-A promotes brain insulin resistance in Alzheimer disease: A new paradigm. Free Radic Biol Med 2016; 91:127-42. [PMID: 26698666 DOI: 10.1016/j.freeradbiomed.2015.12.012] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/10/2015] [Accepted: 12/12/2015] [Indexed: 01/12/2023]
Abstract
Clinical studies suggest a link between peripheral insulin resistance and cognitive dysfunction. Interestingly, post-mortem analyses of Alzheimer disease (AD) subjects demonstrated insulin resistance in the brain proposing a role for cognitive deficits observed in AD. However, the mechanisms responsible for the onset of brain insulin resistance (BIR) need further elucidations. Biliverdin reductase-A (BVR-A) emerged as a unique Ser/Thr/Tyr kinase directly involved in the insulin signaling and represents an up-stream regulator of the insulin signaling cascade. Because we previously demonstrated the oxidative stress (OS)-induced impairment of BVR-A in human AD brain, we hypothesize that BVR-A dysregulation could be associated with the onset of BIR in AD. In the present work, we longitudinally analyze the age-dependent changes of (i) BVR-A protein levels and activation, (ii) total oxidative stress markers levels (PC, HNE, 3-NT) as well as (iii) IR/IRS1 levels and activation in the hippocampus of the triple transgenic model of AD (3xTg-AD) mice. Furthermore, ad hoc experiments have been performed in SH-SY5Y neuroblastoma cells to clarify the molecular mechanism(s) underlying changes observed in mice. Our results show that OS-induced impairment of BVR-A kinase activity is an early event, which starts prior the accumulation of Aβ and tau pathology or the elevation of TNF-α, and that greatly contribute to the onset of BIR along the progression of AD pathology in 3xTg-Ad mice. Based on these evidence we, therefore, propose a new paradigm for which: OS-induced impairment of BVR-A is firstly responsible for a sustained activation of IRS1, which then causes the stimulation of negative feedback mechanisms (i.e. mTOR) aimed to turn-off IRS1 hyper-activity and thus BIR. Similar alterations characterize also the normal aging process in mice, positing BVR-A impairment as a possible bridge in the transition from normal aging to AD.
Collapse
Affiliation(s)
- Eugenio Barone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185 Roma, Italy; Universidad Autónoma de Chile, Instituto de Ciencias Biomédicas, Facultad de Salud, Avenida Pedro de Valdivia 425, Providencia, Santiago, Chile
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185 Roma, Italy
| | - Tommaso Cassano
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122 Foggia, Italy
| | - Andrea Arena
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185 Roma, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185 Roma, Italy
| | - Michele Angelo Lavecchia
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Roma, Piazzale A. Moro 5, 00185 Roma, Italy
| | - Raffaella Coccia
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185 Roma, Italy
| | - D Allan Butterfield
- Department of Chemistry, Markey Cancer Center, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506-0055, USA.
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185 Roma, Italy.
| |
Collapse
|
132
|
Granic A, Davies K, Adamson A, Kirkwood T, Hill TR, Siervo M, Mathers JC, Jagger C. Dietary Patterns High in Red Meat, Potato, Gravy, and Butter Are Associated with Poor Cognitive Functioning but Not with Rate of Cognitive Decline in Very Old Adults. J Nutr 2016; 146:265-74. [PMID: 26740685 PMCID: PMC4725429 DOI: 10.3945/jn.115.216952] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 11/30/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Healthy dietary patterns (DPs) have been linked to better cognition and reduced risk of dementia in older adults, but their role in cognitive functioning and decline in the very old (aged ≥85 y) is unknown. OBJECTIVE We investigated the association between previously established DPs from the Newcastle 85+ Study and global and attention-specific cognition over 5 y. METHODS We followed up with 302 men and 489 women (1921 birth cohort from Northeast United Kingdom) for change in global cognition [measured by the Standardized Mini-Mental State Examination (SMMSE)] over 5 y and attention (assessed by the cognitive drug research attention battery) over 3 y. We used 2-step clustering to derive DPs and mixed models to determine the relation between DPs and cognition in the presence of the dementia susceptibility gene. RESULTS Previously, we characterized 3 DPs that differed in intake of red meat, potato, gravy, and butter and varied with key health measures. When compared with participants in DP1 (high red meat) and DP3 (high butter), participants in DP2 (low meat) had higher SMMSE scores at baseline (P < 0.001) and follow-ups, and better initial attention (P < 0.05). Membership in DP1 and DP3 was associated with overall worse SMMSE scores (β = 0.09, P = 0.01 and β = 0.08, P = 0.02, respectively) than membership in DP2 after adjustment for sociodemographic factors, lifestyle, multimorbidity, and body mass index (BMI). Additional adjustment for apolipoprotein (apoE) ε4 genotype attenuated the association to nonsignificant in women but not in men in DP1 (β = 0.13, P = 0.02). Participants in DP1 and DP3 also had overall worse concentration (β = 0.04, P = 0.002 and β = 0.028, P = 0.03, respectively) and focused attention (β = 0.02, P = 0.01 and β = 0.02, P = 0.03, respectively), irrespective of apoE ε4 genotype, but similar rate of decline in all cognitive measures over time. CONCLUSION DPs high in red meat, potato, gravy (DP1), or butter (DP3) were associated with poor cognition but not with the rate of cognitive decline in very old adults.
Collapse
Affiliation(s)
- Antoneta Granic
- The Newcastle University Institute for Ageing, Institute of Health & Society,
| | - Karen Davies
- The Newcastle University Institute for Ageing,,Institute of Health & Society
| | - Ashley Adamson
- The Newcastle University Institute for Ageing,,Institute of Health & Society,,Human Nutrition Research Centre
| | - Thomas Kirkwood
- The Newcastle University Institute for Ageing,,Institute for Cell and Molecular Biosciences
| | - Tom R Hill
- Human Nutrition Research Centre,,School of Agriculture, Food and Rural Development, and
| | - Mario Siervo
- The Newcastle University Institute for Ageing,,Human Nutrition Research Centre,,Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - John C Mathers
- The Newcastle University Institute for Ageing,,Human Nutrition Research Centre,,Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Carol Jagger
- The Newcastle University Institute for Ageing,,Institute of Health & Society
| |
Collapse
|
133
|
Abner EL, Nelson PT, Kryscio RJ, Schmitt FA, Fardo DW, Woltjer RL, Cairns NJ, Yu L, Dodge HH, Xiong C, Masaki K, Tyas SL, Bennett DA, Schneider JA, Arvanitakis Z. Diabetes is associated with cerebrovascular but not Alzheimer's disease neuropathology. Alzheimers Dement 2016; 12:882-9. [PMID: 26812281 DOI: 10.1016/j.jalz.2015.12.006] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 11/20/2015] [Accepted: 12/07/2015] [Indexed: 01/21/2023]
Abstract
INTRODUCTION The relationship of diabetes to specific neuropathologic causes of dementia is incompletely understood. METHODS We used logistic regression to evaluate the association between diabetes and infarcts, Braak neurofibrillary tangle stage, and neuritic plaque score in 2365 autopsied persons. In a subset of >1300 persons with available cognitive data, we examined the association between diabetes and cognition using Poisson regression. RESULTS Diabetes increased odds of brain infarcts (odds ratio [OR] = 1.57, P < .0001), specifically lacunes (OR = 1.71, P < .0001), but not Alzheimer's disease neuropathology. Diabetes plus infarcts was associated with lower cognitive scores at end of life than infarcts or diabetes alone, and diabetes plus high level of Alzheimer's neuropathologic changes was associated with lower mini-mental state examination scores than the pathology alone. DISCUSSION This study supports the conclusions that diabetes increases the risk of cerebrovascular but not Alzheimer's disease pathology, and at least some of diabetes' relationship to cognitive impairment may be modified by neuropathology.
Collapse
Affiliation(s)
- Erin L Abner
- Sanders-Brown Center on Aging and Alzheimer's Disease Center, University of Kentucky, Lexington, KY, USA; College of Public Health, Department of Epidemiology, University of Kentucky, Lexington, KY, USA; College of Public Health, Department of Biostatistics, University of Kentucky, Lexington, KY, USA.
| | - Peter T Nelson
- Sanders-Brown Center on Aging and Alzheimer's Disease Center, University of Kentucky, Lexington, KY, USA; College of Medicine, Department of Pathology, University of Kentucky, Lexington, KY, USA
| | - Richard J Kryscio
- Sanders-Brown Center on Aging and Alzheimer's Disease Center, University of Kentucky, Lexington, KY, USA; College of Public Health, Department of Biostatistics, University of Kentucky, Lexington, KY, USA
| | - Frederick A Schmitt
- Sanders-Brown Center on Aging and Alzheimer's Disease Center, University of Kentucky, Lexington, KY, USA; College of Medicine, Department of Neurology, University of Kentucky, Lexington, KY, USA
| | - David W Fardo
- Sanders-Brown Center on Aging and Alzheimer's Disease Center, University of Kentucky, Lexington, KY, USA; College of Public Health, Department of Biostatistics, University of Kentucky, Lexington, KY, USA
| | - Randall L Woltjer
- Layton Aging and Alzheimer's Disease Center, Oregon Health & Science University, Portland, OR, USA; School of Medicine, Department of Pathology, Oregon Health & Science University, Portland, OR, USA
| | - Nigel J Cairns
- Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO, USA; School of Medicine, Department of Neurology, Washington University, St. Louis, MO, USA
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Rush University Medical Center, Department of Neurological Sciences, Chicago, IL, USA
| | - Hiroko H Dodge
- Layton Aging and Alzheimer's Disease Center, Oregon Health & Science University, Portland, OR, USA; School of Medicine, Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Chengjie Xiong
- Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO, USA; School of Medicine, Division of Biostatistics, Washington University, St. Louis, MO, USA
| | - Kamal Masaki
- Kuakini Medical Center and John A. Burns School of Medicine, Department of Geriatric Medicine, University of Hawaii, Honolulu, HI, USA
| | - Suzanne L Tyas
- School of Public Health and Health Systems, Department of Psychology, University of Waterloo, Waterloo, ON, Canada
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Rush University Medical Center, Department of Neurological Sciences, Chicago, IL, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Rush University Medical Center, Department of Pathology, Chicago, IL, USA
| | - Zoe Arvanitakis
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Rush University Medical Center, Department of Neurological Sciences, Chicago, IL, USA
| |
Collapse
|
134
|
Reducing Ribosomal Protein S6 Kinase 1 Expression Improves Spatial Memory and Synaptic Plasticity in a Mouse Model of Alzheimer's Disease. J Neurosci 2016; 35:14042-56. [PMID: 26468204 DOI: 10.1523/jneurosci.2781-15.2015] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
UNLABELLED Aging is the most important risk factor associated with Alzheimer's disease (AD); however, the molecular mechanisms linking aging to AD remain unclear. Suppression of the ribosomal protein S6 kinase 1 (S6K1) increases healthspan and lifespan in several organisms, from nematodes to mammals. Here we show that S6K1 expression is upregulated in the brains of AD patients. Using a mouse model of AD, we found that genetic reduction of S6K1 improved synaptic plasticity and spatial memory deficits, and reduced the accumulation of amyloid-β and tau, the two neuropathological hallmarks of AD. Mechanistically, these changes were linked to reduced translation of tau and the β-site amyloid precursor protein cleaving enzyme 1, a key enzyme in the generation of amyloid-β. Our results implicate S6K1 dysregulation as a previously unidentified molecular mechanism underlying synaptic and memory deficits in AD. These findings further suggest that therapeutic manipulation of S6K1 could be a valid approach to mitigate AD pathology. SIGNIFICANCE STATEMENT Aging is the most important risk factor for Alzheimer's disease (AD). However, little is known about how it contributes to AD pathogenesis. S6 kinase 1 (S6K1) is a protein kinase involved in regulation of protein translation. Reducing S6K1 activity increases lifespan and healthspan. We report the novel finding that reducing S6K1 activity in 3xTg-AD mice ameliorates synaptic and cognitive deficits. These improvement were associated with a reduction in amyloid-β and tau pathology. Mechanistically, lowering S6K1 levels reduced translation of β-site amyloid precursor protein cleaving enzyme 1 and tau, two key proteins involved in AD pathogenesis. These data suggest that S6K1 may represent a molecular link between aging and AD. Given that aging is the most important risk factor for most neurodegenerative diseases, our results may have far-reaching implications into other diseases.
Collapse
|
135
|
Meeker KD, Meabon JS, Cook DG. Partial Loss of the Glutamate Transporter GLT-1 Alters Brain Akt and Insulin Signaling in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2016; 45:509-20. [PMID: 25589729 DOI: 10.3233/jad-142304] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The glutamate transporter GLT-1 (also called EAAT2 in humans) plays a critical role in regulating extracellular glutamate levels in the central nervous system (CNS). In Alzheimer's disease (AD), EAAT2 loss is associated with neuropathology and cognitive impairment. In keeping with this, we have reported that partial GLT-1 loss (GLT-1+/-) causes early-occurring cognitive deficits in mice harboring familial AD AβPPswe/PS1ΔE9 mutations. GLT-1 plays important roles in several molecular pathways that regulate brain metabolism, including Akt and insulin signaling in astrocytes. Significantly, AD pathogenesis also involves chronic Akt activation and reduced insulin signaling in the CNS. In this report we tested the hypothesis that GLT-1 heterozygosity (which reduces GLT-1 to levels that are comparable to losses in AD patients) in AβPPswe/PS1ΔE9 mice would induce sustained activation of Akt and disturb components of the CNS insulin signaling cascade. We found that partial GLT-1 loss chronically increased Akt activation (reflected by increased phosphorylation at serine 473), impaired insulin signaling (reflected by decreased IRβ phosphorylation of tyrosines 1150/1151 and increased IRS-1 phosphorylation at serines 632/635 - denoted as 636/639 in humans), and reduced insulin degrading enzyme (IDE) activity in brains of mice expressing familial AβPPswe/PS1ΔE9 AD mutations. GLT-1 loss also caused an apparent compensatory increase in IDE activity in the liver, an organ that has been shown to regulate peripheral amyloid-β levels and expresses GLT-1. Taken together, these findings demonstrate that partial GLT-1 loss can cause insulin/Akt signaling abnormalities that are in keeping with those observed in AD.
Collapse
Affiliation(s)
- Kole D Meeker
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, USA Geriatric Research, Education, and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System (VAPSHCS), Seattle, WA, USA
| | - James S Meabon
- Mental Illness Research, Education, and Clinical Center (MIRECC), Veterans Affairs Puget Sound Health Care System (VAPSHCS), Seattle, WA, USA Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - David G Cook
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, USA Geriatric Research, Education, and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System (VAPSHCS), Seattle, WA, USA Division of Gerontology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
136
|
Groeneveld ON, Kappelle LJ, Biessels GJ. Potentials of incretin-based therapies in dementia and stroke in type 2 diabetes mellitus. J Diabetes Investig 2016; 7:5-16. [PMID: 26816596 PMCID: PMC4718099 DOI: 10.1111/jdi.12420] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 08/12/2015] [Accepted: 08/17/2015] [Indexed: 12/18/2022] Open
Abstract
Patients with type 2 diabetes mellitus are at risk for accelerated cognitive decline and dementia. Furthermore, their risk of stroke is increased and their outcome after stroke is worse than in those without diabetes. Incretin-based therapies are a class of antidiabetic agents that are of interest in relation to these cerebral complications of diabetes. Two classes of incretin-based therapies are currently available: the glucagon-like-peptide-1 agonists and the dipeptidyl peptidase-4 -inhibitors. Independent of their glucose-lowering effects, incretin-based therapies might also have direct or indirect beneficial effects on the brain. In the present review, we discuss the potential of incretin-based therapies in relation to dementia, in particular Alzheimer's disease, and stroke in patients with type 2 diabetes. Experimental studies on Alzheimer's disease have found beneficial effects of incretin-based therapies on cognition, synaptic plasticity and metabolism of amyloid-β and microtubule-associated protein tau. Preclinical studies on incretin-based therapies in stroke have shown an improved functional outcome, a reduction of infarct volume as well as neuroprotective and neurotrophic properties. Both with regard to the treatment of Alzheimer's disease, and with regard to prevention and treatment of stroke, randomized controlled trials in patients with or without diabetes are underway. In conclusion, experimental studies show promising results of incretin-based therapies at improving the outcome of Alzheimer's disease and stroke through glucose-independent pleiotropic effects on the brain. If these findings would indeed be confirmed in large clinical randomized controlled trials, this would have substantial impact.
Collapse
Affiliation(s)
- Onno N Groeneveld
- University Medical Center UtrechtBrain Center Rudolf MagnusDepartment of NeurologyUtrechtthe Netherlands
| | - L Jaap Kappelle
- University Medical Center UtrechtBrain Center Rudolf MagnusDepartment of NeurologyUtrechtthe Netherlands
| | - Geert Jan Biessels
- University Medical Center UtrechtBrain Center Rudolf MagnusDepartment of NeurologyUtrechtthe Netherlands
| |
Collapse
|
137
|
Fadel JR, Reagan LP. Stop signs in hippocampal insulin signaling: the role of insulin resistance in structural, functional and behavioral deficits. Curr Opin Behav Sci 2015; 9:47-54. [PMID: 26955646 DOI: 10.1016/j.cobeha.2015.12.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In peripheral tissues insulin activates signaling cascades to facilitate glucose uptake from the blood into tissues like liver, muscle and fat. While insulin appears to play a minor role in the regulation of glucose uptake in the central nervous system (CNS), insulin is known to play a major role in regulating synaptic plasticity in brain regions like the hippocampus. The concept that insulin regulates hippocampal neuroplasticity is further supported from animal models of type 2 diabetes (T2DM) and Alzheimer's disease (AD). The goal of this review is to provide an overview of these studies, as well as the studies that have examined whether deficits in hippocampal insulin signaling are amenable to intervention strategies.
Collapse
Affiliation(s)
- Jim R Fadel
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Lawrence P Reagan
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA; WJB Dorn Veterans Affairs Medical Center, Columbia, SC, USA
| |
Collapse
|
138
|
Ekblad LL, Rinne JO, Puukka PJ, Laine HK, Ahtiluoto SE, Sulkava RO, Viitanen MH, Jula AM. Insulin resistance is associated with poorer verbal fluency performance in women. Diabetologia 2015; 58:2545-53. [PMID: 26276262 DOI: 10.1007/s00125-015-3715-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/10/2015] [Indexed: 10/23/2022]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes is an independent risk factor for cognitive decline. Insulin resistance occurring during midlife may increase the risk of cognitive decline later in life. We hypothesised that insulin resistance is associated with poorer cognitive performance and that sex and APOE*E4 might modulate this association. METHODS The association of insulin resistance and APOE*E4 genotype on cognitive function was evaluated in a nationwide Finnish population-based study (n = 5,935, mean age 52.5 years, range 30-97 years). HOMA-IR was used to measure insulin resistance. Cognitive function was tested by word-list learning, word-list delayed-recall, categorical verbal fluency and simple and visual-choice reaction-time tests. Linear regression analysis was used to determine the association between HOMA-IR and the results of the cognitive tests. RESULTS Higher HOMA-IR was associated with poorer verbal fluency in women (p < 0.0001) but not in men (p = 0.56). Higher HOMA-IR was also associated with poorer verbal fluency in APOE*E4 -negative individuals (p = 0.0003), but not in APOE*E4 carriers (p = 0.28). Furthermore, higher HOMA-IR was associated with a slower simple reaction time in the whole study group (p = 0.02). CONCLUSIONS/INTERPRETATION To our knowledge, this is the first comprehensive, population-based study, including both young and middle-aged adults, to report that female sex impacts the association of HOMA-IR with verbal fluency. Our study was cross-sectional, so causal effects of HOMA-IR on cognition could not be evaluated. However, our results suggest that HOMA-IR could be an early marker for an increased risk of cognitive decline in women.
Collapse
Affiliation(s)
- Laura L Ekblad
- Turku PET Centre, University of Turku and Turku University Hospital, P.O. Box 52, 20521, Turku, Finland.
- Turku Health Care Centre, Turku, Finland.
| | - Juha O Rinne
- Turku PET Centre, University of Turku and Turku University Hospital, P.O. Box 52, 20521, Turku, Finland
- Division of Clinical Neurosciences, Turku University Hospital, Turku, Finland
| | - Pauli J Puukka
- National Institute for Health and Welfare (THL), Turku, Finland
| | - Hanna K Laine
- Turku City Hospital, University of Turku, Turku, Finland
- Department of Medicine, University of Turku, Turku, Finland
| | | | | | - Matti H Viitanen
- Turku City Hospital, University of Turku, Turku, Finland
- Clinical Geriatrics, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | - Antti M Jula
- National Institute for Health and Welfare (THL), Turku, Finland
| |
Collapse
|
139
|
Abstract
Clinical studies suggest a link between type 2 diabetes mellitus (T2DM) and insulin resistance (IR) and cognitive dysfunction, but there are significant gaps in our knowledge of the mechanisms underlying this relationship. Animal models of IR help to bridge these gaps and point to hippocampal IR as a potential mediator of cognitive dysfunction in T2DM, as well as in Alzheimer disease (AD). This Review highlights these observations and discusses intervention studies which suggest that the restoration of insulin activity in the hippocampus may be an effective strategy to alleviate the cognitive decline associated with T2DM and AD.
Collapse
|
140
|
Frazier DT, Bettcher BM, Dutt S, Patel N, Mungas D, Miller J, Green R, Kramer JH. Relationship between Insulin-Resistance Processing Speed and Specific Executive Function Profiles in Neurologically Intact Older Adults. J Int Neuropsychol Soc 2015; 21:622-8. [PMID: 26272269 PMCID: PMC4764989 DOI: 10.1017/s1355617715000624] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
This study investigated the relationship between insulin-resistance and constituent components of executive function in a sample of neurologically intact older adult subjects using the homeostasis model assessment (HOMA-IR) and latent factors of working memory, cognitive control and processing speed derived from confirmatory factor analysis. Low-density lipoprotein (LDL), mean arterial pressure (MAP), along with body mass index (BMI) and white matter hypointensity (WMH) were used to control for vascular risk factors, adiposity and cerebrovascular injury. The study included 119 elderly subjects recruited from the University of California, San Francisco Memory and Aging Center. Subjects underwent neuropsychological assessment, fasting blood draw and brain magnetic resonance imaging (MRI). Partial correlations and linear regression models were used to examine the HOMA-IR-executive function relationship. Pearson correlation adjusting for age showed a significant relationship between HOMA-IR and working memory (rp = -.18; p = .047), a trend with cognitive control (rp = -.17; p = .068), and no relationship with processing speed (rp = .013; p = .892). Linear regression models adjusting for demographic factors (age, education, and gender), LDL, MAP, BMI, and WMH indicated that HOMA-IR was negatively associated with cognitive control (r = -.256; p = .026) and working memory (r = -.234; p = .054). These results suggest a greater level of peripheral insulin-resistance is associated with decreased cognitive control and working memory. After controlling for demographic factors, vascular risk, adiposity and cerebrovascular injury, HOMA-IR remained significantly associated with cognitive control, with working memory showing a trend. These findings substantiate the insulin-resistance-executive function hypothesis and suggest a complex interaction, demonstrated by the differential impact of insulin-resistance on processing speed and specific aspects of executive function.
Collapse
Affiliation(s)
- Darvis T. Frazier
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA 94143
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143
| | - Brianne M. Bettcher
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA 94143
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143
| | - Shubir Dutt
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA 94143
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143
| | - Nihar Patel
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA 94143
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143
| | - Dan Mungas
- Department of Neurology, School of Medicine, University of California, Davis, Davis, CA 95817
| | - Joshua Miller
- Department of Pathology and Laboratory Medicine, University of California, Davis, Davis, CA 95817
| | - Ralph Green
- Department of Pathology and Laboratory Medicine, University of California, Davis, Davis, CA 95817
| | - Joel H. Kramer
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA 94143
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143
| |
Collapse
|
141
|
Tikhonoff V, Casiglia E, Guidotti F, Giordano N, Martini B, Mazza A, Spinella P, Palatini P. Body fat and the cognitive pattern: A population-based study. Obesity (Silver Spring) 2015; 23:1502-10. [PMID: 26110893 DOI: 10.1002/oby.21114] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 03/13/2015] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The relationship between body fatness and cognitive pattern at a population level was investigated. METHODS Among 500 unselected subjects from the general population, the role of body mass index (BMI) and body fat mass (BFM) on a mini-mental state examination (MMSE) and on a battery of paper and pencil neuropsychological tests was analyzed. Multiple linear regressions, accounting for potential confounders, were used. RESULTS In fully adjusted models, MMSE (coefficient +0.027, 95% confidence intervals, 0.017-0.177), the clock drawing test (+0.141, 0.053-0.226), and the trail making test A (+1.542, 0.478-2.607) were positively associated with BMI. Adding BFM to the models, no associations were observed. The tests were also positively associated with BFM (+0.056, 0.021-0.091; +0.063, 0.025-0.101; +0.592, 0.107-1.077; respectively). At analysis of covariance, the same tests were significantly better performed over 29.4 kg m(-2) of BMI. After adding BFM as further confounder, all differences in performance across BMI were no longer significant. The three tests were better performed over 34.6 kg of BFM. CONCLUSIONS Higher BMI and particularly higher BFM are positively associated with better performance at the cognitive tasks exploring selective attention and executive functions.
Collapse
Affiliation(s)
- Valérie Tikhonoff
- Department of Medicine, University of Padova, Padova, Italy
- MRC Unit for Lifelong Health and Ageing at UCL, University College of London, London, UK
| | | | | | | | - Bortolo Martini
- Department of Cardiology, Hospital of Santorso, Thiene, Italy
| | - Alberto Mazza
- Department of Medicine, Hospital of Rovigo, Rovigo, Italy
| | - Paolo Spinella
- Department of Medicine, University of Padova, Padova, Italy
| | - Paolo Palatini
- Department of Medicine, University of Padova, Padova, Italy
| |
Collapse
|
142
|
Lioutas VA, Alfaro-Martinez F, Bedoya F, Chung CC, Pimentel DA, Novak V. Intranasal Insulin and Insulin-Like Growth Factor 1 as Neuroprotectants in Acute Ischemic Stroke. Transl Stroke Res 2015; 6:264-75. [PMID: 26040423 DOI: 10.1007/s12975-015-0409-7] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 04/16/2015] [Accepted: 05/13/2015] [Indexed: 12/22/2022]
Abstract
Treatment options for stroke remain limited. Neuroprotective therapies, in particular, have invariably failed to yield the expected benefit in stroke patients, despite robust theoretical and mechanistic background and promising animal data. Insulin and insulin-like growth factor 1 (IGF-1) play a pivotal role in critical brain functions, such as energy homeostasis, neuronal growth, and differentiation. They may exhibit neuroprotective properties in acute ischemic stroke based upon their vasodilatory, anti-inflammatory and antithrombotic effects, as well as improvements of functional connectivity, neuronal metabolism, neurotransmitter regulation, and remyelination. Intranasally administered insulin has demonstrated a benefit for prevention of cognitive decline in older people, and IGF-1 has shown potential benefit to improve functional outcomes in animal models of acute ischemic stroke. The intranasal route presents a feasible, tolerable, safe, and particularly effective administration route, bypassing the blood-brain barrier and maximizing distribution to the central nervous system (CNS), without the disadvantages of systemic side effects and first-pass metabolism. This review summarizes the neuroprotective potential of intranasally administered insulin and IGF-1 in stroke patients. We present the theoretical background and pathophysiologic mechanisms, animal and human studies of intranasal insulin and IGF-1, and the safety and feasibility of intranasal route for medication administration to the CNS.
Collapse
Affiliation(s)
- Vasileios-Arsenios Lioutas
- Department of Neurology, Division of Cerebrovascular Diseases, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Palmer 127, Boston, MA, 02215, USA,
| | | | | | | | | | | |
Collapse
|
143
|
Yang CM, Shen YC, Weng SF, Wang JJ, Tien KJ. Increased Risk of Dementia in Patients With Erectile Dysfunction: A Population-Based, Propensity Score-Matched, Longitudinal Follow-Up Study. Medicine (Baltimore) 2015; 94:e990. [PMID: 26091478 PMCID: PMC4616558 DOI: 10.1097/md.0000000000000990] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Erectile dysfunction (ED) is a well-known predictor for future cardiovascular and cerebrovascular disease. However, the relationship between ED and dementia has rarely been examined. This study investigates the longitudinal risk for Alzheimer's disease and non-Alzheimer dementia in patients with ED. We collected a random sample of 1,000,000 individuals from Taiwan's National Health Insurance database. From this sample, we identified 4153 patients with newly diagnosed ED between 2000 and 2009 and compared them with a matched cohort of 20,765 patients without ED. All patients were tracked for 7 years from the index date to identify which of them subsequently developed dementia. During the 7-year follow-up period, the incidence rate of dementia in the ED cohort was 35.33 per 10,000 person-years. In the comparison groups, it was 21.67 per 10,000 person-years. After adjustment for patients characteristics and comorbidities, patients with ED were 1.68-times more likely to develop dementia than patients without ED (95% CI = 1.34-2.10, P < 0.0001). In addition, older patients and those with diabetes, hypertension, chronic kidney disease, stroke, depression, and anxiety were found to be at increased risk for dementia. Analyzing the data by dementia type, we found the hazard risk for Alzheimer's disease and non-Alzheimer dementia to be greater in patients with ED (adjusted HR 1.68, 95% CI = 1.31-2.16, P < 0.0001 and 1.63, 95% CI = 1.02-2.62, P = 0.0429, respectively). Log-rank test revealed that patients with ED had significantly higher cumulative incidence rates of dementia than those without (P < 0.0001). Patients with ED are at an increased risk for dementia later in life.
Collapse
Affiliation(s)
- Chun-Ming Yang
- From the Department of Neurology, Chi Mei Medical Center, Tainan (C-MY); Department of Urology, Kaohsiung Chang Gung Memorial Hospital (Y-CS); Cheng Shiu University, Kaohsiung (Y-CS); Department of Medical Research, Chi Mei Medical Center (S-FW, J-JW); Department of Hospital and Health Care Administration, Chia Nan University of Pharmacy and Science (S-FW); Division of Endocrinology and Metabolism, Department of Internal Medicine, Chi Mei Medical Center (K-JT); and Department of Senior Citizen Service Management, Chia Nan University of Pharmacy and Science, Tainan, Taiwan (K-JT)
| | | | | | | | | |
Collapse
|
144
|
Herrup K. The case for rejecting the amyloid cascade hypothesis. Nat Neurosci 2015; 18:794-9. [PMID: 26007212 DOI: 10.1038/nn.4017] [Citation(s) in RCA: 537] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 04/09/2015] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a biologically complex neurodegenerative dementia. Nearly 20 years ago, with the combination of observations from biochemistry, neuropathology and genetics, a compelling hypothesis known as the amyloid cascade hypothesis was formulated. The core of this hypothesis is that it is pathological accumulations of amyloid-β, a peptide fragment of a membrane protein called amyloid precursor protein, that act as the root cause of AD and initiate its pathogenesis. Yet, with the passage of time, growing amounts of data have accumulated that are inconsistent with the basically linear structure of this hypothesis. And while there is fear in the field over the consequences of rejecting it outright, clinging to an inaccurate disease model is the option we should fear most. This Perspective explores the proposition that we are over-reliant on amyloid to define and diagnose AD and that the time has come to face our fears and reject the amyloid cascade hypothesis.
Collapse
Affiliation(s)
- Karl Herrup
- 1] Division of Life Science, Hong Kong University of Science and Technology, Kowloon, Hong Kong. [2] State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Kowloon, Hong Kong
| |
Collapse
|
145
|
Hansell NK, Halford GS, Andrews G, Shum DHK, Harris SE, Davies G, Franic S, Christoforou A, Zietsch B, Painter J, Medland SE, Ehli EA, Davies GE, Steen VM, Lundervold AJ, Reinvang I, Montgomery GW, Espeseth T, Hulshoff Pol HE, Starr JM, Martin NG, Le Hellard S, Boomsma DI, Deary IJ, Wright MJ. Genetic basis of a cognitive complexity metric. PLoS One 2015; 10:e0123886. [PMID: 25860228 PMCID: PMC4393228 DOI: 10.1371/journal.pone.0123886] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 02/23/2015] [Indexed: 01/15/2023] Open
Abstract
Relational complexity (RC) is a metric reflecting capacity limitation in relational processing. It plays a crucial role in higher cognitive processes and is an endophenotype for several disorders. However, the genetic underpinnings of complex relational processing have not been investigated. Using the classical twin model, we estimated the heritability of RC and genetic overlap with intelligence (IQ), reasoning, and working memory in a twin and sibling sample aged 15-29 years (N = 787). Further, in an exploratory search for genetic loci contributing to RC, we examined associated genetic markers and genes in our Discovery sample and selected loci for replication in four independent samples (ALSPAC, LBC1936, NTR, NCNG), followed by meta-analysis (N>6500) at the single marker level. Twin modelling showed RC is highly heritable (67%), has considerable genetic overlap with IQ (59%), and is a major component of genetic covariation between reasoning and working memory (72%). At the molecular level, we found preliminary support for four single-marker loci (one in the gene DGKB), and at a gene-based level for the NPS gene, having influence on cognition. These results indicate that genetic sources influencing relational processing are a key component of the genetic architecture of broader cognitive abilities. Further, they suggest a genetic cascade, whereby genetic factors influencing capacity limitation in relational processing have a flow-on effect to more complex cognitive traits, including reasoning and working memory, and ultimately, IQ.
Collapse
Affiliation(s)
- Narelle K. Hansell
- Neuroimaging Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- * E-mail:
| | - Graeme S. Halford
- School of Applied Psychology, Griffith University, Mt Gravatt Campus, Brisbane, Australia
- Behavioural Basis of Health Program, Griffith Health Institute and School of Applied Psychology, Griffith University, Brisbane, Australia
| | - Glenda Andrews
- Behavioural Basis of Health Program, Griffith Health Institute and School of Applied Psychology, Griffith University, Brisbane, Australia
- School of Applied Psychology, Griffith University, Gold Coast Campus, Southport, Australia
| | - David H. K. Shum
- Behavioural Basis of Health Program, Griffith Health Institute and School of Applied Psychology, Griffith University, Brisbane, Australia
| | - Sarah E. Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Gail Davies
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, United Kingdom
- Department of Psychology, University of Edinburgh, Edinburgh, United Kingdom
| | - Sanja Franic
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Andrea Christoforou
- K.G. Jebsen Centre for Psychosis Research and the Norwegian Center for Mental Disorders Research (NORMENT), Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Brendan Zietsch
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- School of Psychology, University of Queensland, St Lucia, Brisbane, Australia
| | - Jodie Painter
- Molecular Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Sarah E. Medland
- Quantitative Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Erik A. Ehli
- Avera Institute for Human Genetics, Avera McKennan Hospital & University Health Center, Sioux Falls, South Dakota, United States of America
| | - Gareth E. Davies
- Avera Institute for Human Genetics, Avera McKennan Hospital & University Health Center, Sioux Falls, South Dakota, United States of America
| | - Vidar M. Steen
- K.G. Jebsen Centre for Psychosis Research and the Norwegian Center for Mental Disorders Research (NORMENT), Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Astri J. Lundervold
- K.G. Jebsen Center for Research on Neuropsychiatric Disorders, University of Bergen, Bergen, Norway
- Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
- Center for Research on Aging and Dementia, Haraldsplass Deaconess Hospital, Bergen, Norway
| | - Ivar Reinvang
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Grant W. Montgomery
- Molecular Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Thomas Espeseth
- Department of Psychology, University of Oslo, Oslo, Norway
- Norwegian Center for Mental Disorders Research (NORMENT) and the K.G. Jebsen Center for Psychosis Research, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Hilleke E. Hulshoff Pol
- Brain Center Rudolf Magnus, Department of Psychiatry, University Medical Center Utrecht, Utrecht, The Netherlands
| | - John M. Starr
- Department of Psychology, University of Edinburgh, Edinburgh, United Kingdom
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Nicholas G. Martin
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Stephanie Le Hellard
- K.G. Jebsen Centre for Psychosis Research and the Norwegian Center for Mental Disorders Research (NORMENT), Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Dorret I. Boomsma
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Ian J. Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Margaret J. Wright
- Neuroimaging Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| |
Collapse
|
146
|
Affiliation(s)
- A E Roher
- Banner Sun Health Research Institute, Sun City, AZ, USA
| |
Collapse
|
147
|
Humanin Does Not Protect Against STZ-Induced Spatial Memory Impairment. J Mol Neurosci 2015; 56:290-8. [PMID: 25744099 DOI: 10.1007/s12031-015-0531-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 02/17/2015] [Indexed: 01/05/2023]
Abstract
[Gly14]-Humanin (HNG) is a 24-amino acid peptide which was first identified in the brains of patients diagnosed with Alzheimer's disease (AD). In this region, some neurons were protected against cell damage occurring in this disease. Further studies suggested a neuroprotective role for humanin against Aβ and some other insults. Intraventricularly administered streptozotocin (STZ) disrupts insulin signaling pathway which leads to behavioral and biochemical changes resemble to early signs of AD; therefore, STZ model has been proposed as a model for sporadic Alzheimer's disease (sAD). Regarding the reported beneficial effects of humanin in AD, this study was aimed to investigate if this peptide prevents spatial memory and hippocampal PI3/Akt signaling impairment induced by centrally injected STZ. Adult male Sprague-Dawely rats weighting 250-300 g were used, and cannuls were implanted bilaterally into lateral ventricles. STZ was administered on days 1 and 3 (3 mg/kg), and humanin (0.01, 0.05, 0.1, and 1 nmol) or saline were injected from day 4 and continued till day 14. The animal's learning and memory capability was assessed on days 15-18 using Morris water maze. After complement of behavioral studies, the hippocampi were isolated, and the level of phosphorylated Akt (pAkt) was assessed through Western blot analysis. The results showed that STZ significantly impaired spatial memory, and humanin in a wide range of doses (0.01, 0.05, 0.1, and 1 nmol) failed to restore STZ-induced deficit. It was also revealed that humanin was not efficient in restoring pAkt disruption. It seems that humanin is not capable in restoring memory deterioration that resulted from insulin signaling disruption.
Collapse
|
148
|
Barone E, Butterfield DA. Insulin resistance in Alzheimer disease: Is heme oxygenase-1 an Achille's heel? Neurobiol Dis 2015; 84:69-77. [PMID: 25731746 DOI: 10.1016/j.nbd.2015.02.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 02/18/2015] [Indexed: 01/10/2023] Open
Abstract
Insulin resistance, clinically defined as the inability of insulin to increase glucose uptake and utilization, has been found to be associated with the progression of Alzheimer disease (AD). Indeed, postmortem AD brain shows all the signs of insulin resistance including: (i) reduced brain insulin receptor (IR) sensitivity, (ii) hypophosphorylation of the insulin receptor and downstream second messengers such as IRS-1, and (iii) attenuated insulin and insulin growth factor (IGF)-1 receptor expression. However, the exact mechanisms driving insulin resistance have not been completely elucidated. Quite recently, the levels of the peripheral inducible isoform of heme oxygenase (HO-1), a well-known protein up-regulated during cell stress response, were proposed to be among the strongest positive predictors of metabolic disease, including insulin resistance. Because our group previously reported on levels, activation state and oxidative stress-induced post-translational modifications of HO-1 in AD brain and our ongoing studies to better elucidate the role of HO-1 in insulin resistance-associated AD pathology, the aim of this review is to provide reader with a critical analysis on new aspects of the interplay between HO-1 and insulin resistance and on how the available lines of evidence could be useful for further comprehension of processes in AD brain.
Collapse
Affiliation(s)
- Eugenio Barone
- Department of Biochemical Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506-0055, USA.
| |
Collapse
|
149
|
Koekkoek PS, Kappelle LJ, van den Berg E, Rutten GEHM, Biessels GJ. Cognitive function in patients with diabetes mellitus: guidance for daily care. Lancet Neurol 2015; 14:329-40. [PMID: 25728442 DOI: 10.1016/s1474-4422(14)70249-2] [Citation(s) in RCA: 257] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Diabetes mellitus is associated with an increase in the risk of dementia and the proportion of patients who convert from mild cognitive impairment (MCI) to dementia. In addition to MCI and dementia, the stages of diabetes-associated cognitive dysfunction include subtle cognitive changes that are unlikely to affect activities of daily life or diabetes self-management. These diabetes-associated cognitive decrements have structural brain correlates detectable with brain MRI, but usually show little progression over time. Although cognitive decrements do not generally represent a pre-dementia stage in patients below the age of 60-65 years, in older individuals these subtle cognitive changes might represent the earliest stages of a dementia process. Acknowledgment of diabetes-associated cognitive decrements can help to improve understanding of patients' symptoms and guide management. Future challenges are to establish the importance of screening for cognitive impairment in people with diabetes, to identify those at increased risk of accelerated cognitive decline at an early stage, and to develop effective treatments.
Collapse
Affiliation(s)
- Paula S Koekkoek
- Julius Center for Health Sciences and Primary Care, University Medical Centre Utrecht, Netherlands
| | - L Jaap Kappelle
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Centre Utrecht, Netherlands.
| | - Esther van den Berg
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Centre Utrecht, Netherlands; Experimental Psychology, Helmholtz Institute, Utrecht University, Utrecht, Netherlands
| | - Guy E H M Rutten
- Julius Center for Health Sciences and Primary Care, University Medical Centre Utrecht, Netherlands
| | - Geert Jan Biessels
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Centre Utrecht, Netherlands
| |
Collapse
|
150
|
Caldwell CC, Yao J, Brinton RD. Targeting the prodromal stage of Alzheimer's disease: bioenergetic and mitochondrial opportunities. Neurotherapeutics 2015; 12:66-80. [PMID: 25534394 PMCID: PMC4322082 DOI: 10.1007/s13311-014-0324-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) has a complex and progressive neurodegenerative phenotype, with hypometabolism and impaired mitochondrial bioenergetics among the earliest pathogenic events. Bioenergetic deficits are well documented in preclinical models of mammalian aging and AD, emerge early in the prodromal phase of AD, and in those at risk for AD. This review discusses the importance of early therapeutic intervention during the prodromal stage that precedes irreversible degeneration in AD. Mechanisms of action for current mitochondrial and bioenergetic therapeutics for AD broadly fall into the following categories: 1) glucose metabolism and substrate supply; 2) mitochondrial enhancers to potentiate energy production; 3) antioxidants to scavenge reactive oxygen species and reduce oxidative damage; 4) candidates that target apoptotic and mitophagy pathways to either remove damaged mitochondria or prevent neuronal death. Thus far, mitochondrial therapeutic strategies have shown promise at the preclinical stage but have had little-to-no success in clinical trials. Lessons learned from preclinical and clinical therapeutic studies are discussed. Understanding the bioenergetic adaptations that occur during aging and AD led us to focus on a systems biology approach that targets the bioenergetic system rather than a single component of this system. Bioenergetic system-level therapeutics personalized to bioenergetic phenotype would target bioenergetic deficits across the prodromal and clinical stages to prevent and delay progression of AD.
Collapse
Affiliation(s)
- Charles C. Caldwell
- />Clinical and Experimental Therapeutics Program, School of Pharmacy, University of Southern California, Los Angeles, CA 90089 USA
| | - Jia Yao
- />Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089 USA
| | - Roberta Diaz Brinton
- />Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089 USA
- />Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089 USA
| |
Collapse
|