1
|
Iban-Arias R, Portela ASD, Masieri S, Radu A, Yang EJ, Chen LC, Gordon T, Pasinetti GM. Role of acute exposure to environmental stressors in the gut-brain-periphery axis in the presence of cognitive resilience. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167760. [PMID: 40037471 DOI: 10.1016/j.bbadis.2025.167760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/19/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
Climate change-induced environmental stressors, including ambient particulate matter (PM2.5) and extreme heat stress (HS), pose serious health risks, particularly for neurodegenerative diseases. PM2.5 exacerbates cardiovascular and neurodegenerative conditions, while HS increases mortality and worsens air pollution. Combined exposure may amplify these effects, especially in vulnerable populations at risk for Alzheimer's disease (AD). In our experimental study using a mouse model of early-onset Alzheimer's disease (EOAD), we explored the combined effects of extreme weather conditions, particularly exposure to ambient PM2.5 and HS. Our research indicated that even short, repeated exposure to these environmental stressors disrupts brain energy metabolism and mitochondrial respiratory functions, which we found to be associated with altered hippocampal synaptic functions. Additionally, we find that key mechanisms associated with impaired intestinal permeability and gut dysbiosis are affected, supporting the hypothesis that exposure to climate change communication may also disrupt the gut-brain axis, as in part evidenced in our study by peripheral changes in immune and inflammatory signaling. Moreover, despite significant disruptions in metabolic and immune-inflammatory pathways, we observed no acceleration of cognitive decline in the young asymptomatic EOAD mice subjected to short, repeated exposure to extreme heat and environmental PM2.5. These findings highlight the potential role of climate change in promoting risk factors like neuroinflammation and gut-brain axis dysfunction due to gut microbiome dysbiosis in the onset and progression of AD, particularly in asymptomatic individuals at risk for developing the condition.
Collapse
Affiliation(s)
- Ruth Iban-Arias
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10019, United States of America
| | - Ariana Soares Dias Portela
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10019, United States of America
| | - Sibilla Masieri
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10019, United States of America
| | - Aurelian Radu
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10019, United States of America
| | - Eun-Jeong Yang
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10019, United States of America
| | - Lung-Chi Chen
- Department of Medicine, NYU Langone School of Medicine, New York, NY, 10010, United States of America
| | - Terry Gordon
- Department of Medicine, NYU Langone School of Medicine, New York, NY, 10010, United States of America
| | - Giulio Maria Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10019, United States of America; Geriatrics Research, Education and Clinical Center, JJ Peters VA Medical Center, Bronx, NY, 10468, United States of America.
| |
Collapse
|
2
|
Li L, Li C, Zhu J. The relationship between estimated glucose disposal rate and cognitive function in older individuals. Sci Rep 2025; 15:5874. [PMID: 39966445 PMCID: PMC11836112 DOI: 10.1038/s41598-025-89623-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 02/06/2025] [Indexed: 02/20/2025] Open
Abstract
The estimated glucose disposal rate (eGDR) serves as a novel indicator of insulin resistance, which has been shown to correlate with cardiovascular disease risk; however, its relationship with cognitive function remains unclear.This article describes a cross-sectional study design based on data from the 2011-2014 National Health and Nutrition Examination Survey (NHANES). The Weighted logistic regression and the restricted cubic spline were employed to examine the relationship between eGDR and cognitive ability.The subjects were divided into two categories: the normal group and the cognitive function decline (CFD) group, based on their cognitive scores. There were significant differences in eGDR levels between the two groups(P = 0.001).After adjusting for relevant covariates, notable differences were found between eGDR and cognitive function when eGDR was expressed in both continuous and categorical data forms (P < 0.05). The stability of these findings was further confirmed through sensitivity analyses.This difference persisted in subgroups, including women, individuals with education beyond high school, moderate drinkers, and those who had not been diagnosed with stroke (P < 0.05). A restricted cubic spline revealed a non-linear relationship with an inflection point between the two (P-for-non-linear < 0.05, P-overall < 0.001). This study contributes to the understanding of the relationship between eGDR and cognitive performance by identifying a potential non-linear association.
Collapse
Affiliation(s)
- Li Li
- Department of Neurology, Affiliated Hospital of Chengde Medical College, Hebei, China.
| | - Chengbo Li
- Department of Neurology, Affiliated Hospital of Chengde Medical College, Hebei, China
| | - Jiang Zhu
- Department of Neurology, Affiliated Hospital of Chengde Medical College, Hebei, China
| |
Collapse
|
3
|
Wang P, Wang M, Xie Z, Zhi Z, Wang Y, Liu F, Liu Y, Zhao L. Association Between Four Non-Insulin-Based Insulin Resistance Indices and the Risk of Post-Stroke Depression. Clin Interv Aging 2025; 20:19-31. [PMID: 39817260 PMCID: PMC11733171 DOI: 10.2147/cia.s501569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/30/2024] [Indexed: 01/18/2025] Open
Abstract
Purpose Research suggests that insulin resistance (IR) is associated with acute ischemic stroke (AIS) and depression. The use of insulin-based IR assessments is complicated. Therefore, we explored the relationship between four non-insulin-based IR indices and post-stroke depression (PSD). Patients and Methods A total of 638 consecutive AIS patients were enrolled in this prospective cohort study. Clinical data were collected to compute indices such as the triglyceride glucose (TyG) index, triglyceride glucose-body mass index (TyG-BMI), insulin resistance metabolic score (METS-IR), and triglyceride/high-density lipoprotein cholesterol ratio (TG/HDL-C). One month post-stroke, neuropsychological assessments were conducted using the 17-item Hamilton Depression Scale. Binary logistic regression analysis was performed to explore the relationship between the four non-insulin-based IR indices and PSD. Results Ultimately, 381 patients completed the 1-month follow-up, including 112 (29.4%) with PSD. The PSD group exhibited significantly higher levels of the four IR indices compared to the non-PSD group. Logistic regression analysis demonstrated that these indicators were independently associated with PSD occurrence, both before and after adjusting for potential confounders (all P < 0.001). Tertile analyses indicated that the highest tertile group had a greater risk of PSD occurrence than the lowest tertile group for four IR indicators, even after adjusting for potential confounders (all P < 0.05). Restricted cubic spline analysis revealed a linear dose-response relationship between the four IR indices and PSD. In the subgroup analysis, only the TyG index showed a significant interaction with diabetes (P for interaction = 0.014). The area under curve values for the TyG index, TyG-BMI, METS-IR, and TG/HDL-C were 0.700, 0.721, 0.711, and 0.690, respectively. Conclusion High TyG index, TyG-BMI, METS-IR, and TG/HDL-C at baseline were independent risk factors for PSD in AIS. Each of these indicators exhibits predictive value for PSD occurrence, aiding in the early identification of high-risk groups.
Collapse
Affiliation(s)
- Ping Wang
- Department of Neurology, the Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China
| | - Mengchao Wang
- Department of Neurology, the Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China
| | - Zhe Xie
- Department of Neurology, the Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China
| | - Zhongwen Zhi
- Department of Neurology, the Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China
| | - Yuqian Wang
- Department of Neurology, the Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China
| | - Fan Liu
- Department of Neurology, the Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China
| | - Yufeng Liu
- Department of Neurology, the Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China
| | - Liandong Zhao
- Department of Neurology, the Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China
| |
Collapse
|
4
|
Ghondaghsaz E, Khalaji A, Mahalleh M, Masrour M, Mohammadi P, Cannavo A, Behnoush AH. Exploring the Association Between Cognitive Decline and Triglyceride-Glucose Index: A Systematic Review and Meta-Analysis. Brain Behav 2024; 14:e70131. [PMID: 39482852 PMCID: PMC11527841 DOI: 10.1002/brb3.70131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/21/2024] [Accepted: 10/18/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Cognitive decline and dementia are debilitating conditions that compromise the quality of life and charge the healthcare system with a substantial socioeconomic burden. In this context, emerging evidence supports an association between the triglyceride-glucose index (TyG), a surrogate insulin resistance marker, and cognitive decline and dementia. Hence, we systematically reviewed the studies assessing the TyG index in patients with cognitive decline and their controls. METHODS Online international databases (PubMed, Scopus, Embase, and the Web of Science) were searched comprehensively for studies showing the TyG index in patients with cognitive decline/impairment. Random-effect meta-analyses were conducted to calculate the standardized mean difference (SMD), pooled odds ratio (OR), and pooled area under the curve (AUC), in addition to 95% confidence intervals (CIs) for the comparisons of groups. RESULTS Seventeen studies were included in our analysis. Then, we conducted a meta-analysis, demonstrating that patients with cognitive decline had significantly higher levels of TyG index than those without (SMD 0.83, 95% CI 0.16 to 1.50, p = 0.015). Moreover, our data showed that a 1-unit increase in the TyG index was associated with higher odds of cognitive decline (adjusted OR [aOR] 2.86, 95% CI 1.49 to 5.50, p = 0.002). Further, we observed that patients in the fourth TyG quartile with higher values of the TyG index than the first quartile presented with more increased cognitive decline (aOR 1.62, 95%CI 1.11 to 2.38, p = 0.013). Finally, pooled AUC data for the diagnostic performance of the TyG index resulted in an overall AUC value of 0.73 (95% CI 0.66 to 0.79). Sensitivity and specificity were also calculated as 0.695 and 0.687, respectively. CONCLUSION This study supports the clinical utility of the TyG index in patients with cognitive decline and solicits more focused studies to consolidate its usage in clinical settings and real-world practice.
Collapse
Affiliation(s)
- Elina Ghondaghsaz
- Undergraduate Program in NeuroscienceUniversity of British ColumbiaBritish ColumbiaVancouverCanada
| | - Amirmohammad Khalaji
- School of MedicineTehran University of Medical SciencesTehranIran
- Non‐Communicable Diseases Research CenterEndocrinology and Metabolism Population Sciences InstituteTehran University of Medical SciencesTehranIran
| | - Mehrdad Mahalleh
- School of MedicineTehran University of Medical SciencesTehranIran
- Rheumatology Research CenterTehran University of Medical SciencesTehranIran
| | - Mahdi Masrour
- School of MedicineTehran University of Medical SciencesTehranIran
| | - Parsa Mohammadi
- School of MedicineTehran University of Medical SciencesTehranIran
| | - Alessandro Cannavo
- Department of Translational Medicine SciencesFederico II University of NaplesNaplesItaly
| | - Amir Hossein Behnoush
- School of MedicineTehran University of Medical SciencesTehranIran
- Non‐Communicable Diseases Research CenterEndocrinology and Metabolism Population Sciences InstituteTehran University of Medical SciencesTehranIran
| |
Collapse
|
5
|
Shen J, Wang X, Wang M, Zhang H. Potential molecular mechanism of exercise reversing insulin resistance and improving neurodegenerative diseases. Front Physiol 2024; 15:1337442. [PMID: 38818523 PMCID: PMC11137309 DOI: 10.3389/fphys.2024.1337442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 04/29/2024] [Indexed: 06/01/2024] Open
Abstract
Neurodegenerative diseases are debilitating nervous system disorders attributed to various conditions such as body aging, gene mutations, genetic factors, and immune system disorders. Prominent neurodegenerative diseases include Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and multiple sclerosis. Insulin resistance refers to the inability of the peripheral and central tissues of the body to respond to insulin and effectively regulate blood sugar levels. Insulin resistance has been observed in various neurodegenerative diseases and has been suggested to induce the occurrence, development, and exacerbation of neurodegenerative diseases. Furthermore, an increasing number of studies have suggested that reversing insulin resistance may be a critical intervention for the treatment of neurodegenerative diseases. Among the numerous measures available to improve insulin sensitivity, exercise is a widely accepted strategy due to its convenience, affordability, and significant impact on increasing insulin sensitivity. This review examines the association between neurodegenerative diseases and insulin resistance and highlights the molecular mechanisms by which exercise can reverse insulin resistance under these conditions. The focus was on regulating insulin resistance through exercise and providing practical ideas and suggestions for future research focused on exercise-induced insulin sensitivity in the context of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jiawen Shen
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Xianping Wang
- School of Medicine, Taizhou University, Taizhou, China
| | - Minghui Wang
- College of Sports Medicine, Wuhan Sports University, Wuhan, China
| | - Hu Zhang
- College of Sports Medicine, Wuhan Sports University, Wuhan, China
| |
Collapse
|
6
|
Tian N, Song L, Hou T, Fa W, Dong Y, Liu R, Ren Y, Liu C, Zhu M, Zhang H, Wang Y, Cong L, Du Y, Qiu C. Association of Triglyceride-Glucose Index With Cognitive Function and Brain Atrophy: A Population-Based Study. Am J Geriatr Psychiatry 2024; 32:151-162. [PMID: 37827915 DOI: 10.1016/j.jagp.2023.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 10/14/2023]
Abstract
OBJECTIVE To investigate the associations of triglyceride-glucose (TyG) index, a reliable surrogate marker for insulin resistance, with the function of various cognitive domains and brain structures among older adults. DESIGN A population-based cross-sectional study. SETTING Older adults living in the rural communities in China. PARTICIPANTS About 4,541 rural-dwelling dementia-free participants (age ≥65 years; 56.37% women) undertook examinations in March-September 2018 for MIND-China. MEASUREMENTS TyG index was calculated as ln[fasting triglyceride (mg/dL) × fasting glucose (mg/dL)/2]. A neuropsychological test battery was used to assess memory, attention, verbal fluency, and executive function. Volumetric brain measures were assessed on magnetic resonance imaging (MRI) in a subsample (n = 1,019). Data were analyzed with restricted cubic spline and multivariable general linear models. RESULTS An inverted J-shaped association was observed between TyG index and z-scores of multiple cognitive domains, such that among individuals with TyG index ≥8.57 (median), a higher TyG index was significantly associated with lower z-scores of memory, attention, verbal fluency, executive function, and global cognition (all p < 0.05); among people with TyG index <8.57, a higher TyG index was significantly associated with a higher executive function z-score (p < 0.05), but not with any of the other examined cognitive domains. In the MRI subsample, a higher TyG index was significantly associated with lower volumes of total brain tissue, gray matter, and white matter as well as greater cerebrospinal fluid volume (p < 0.05), but not with white matter hyperintensity volume. CONCLUSIONS Insulin resistance, as indicated by a high TyG index, was associated with poor function in multiple cognitive domains and global brain atrophy.
Collapse
Affiliation(s)
- Na Tian
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China; Medical Science and Technology Innovation Center (NT, YD), Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Lin Song
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China
| | - Tingting Hou
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China
| | - Wenxin Fa
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Yi Dong
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China
| | - Rui Liu
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China
| | - Yifei Ren
- Department of Neurology (YR, YD), Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, P.R. China
| | - Cuicui Liu
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China
| | - Min Zhu
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China
| | - Heng Zhang
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China
| | - Yongxiang Wang
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China; Institute of Brain Science and Brain-Inspired Research (YW, YD), Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China; Aging Research Center and Center for Alzheimer Research (YW, CQ), Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet-Stockholm University, 17165 Solna, Sweden
| | - Lin Cong
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China
| | - Yifeng Du
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Shandong Provincial Clinical Research Center for Neurological Diseases (NT, LS, TH, YD, RL, CL, MZ, HZ, YW, LC, YD), Jinan, Shandong 250021, P.R. China; Medical Science and Technology Innovation Center (NT, YD), Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China; Department of Neurology (YR, YD), Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, P.R. China; Institute of Brain Science and Brain-Inspired Research (YW, YD), Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China.
| | - Chengxuan Qiu
- Department of Neurology (NT, LS, TH, WF, YD, RL, CL, MZ, HZ, YW, LC, YD, CQ), Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China; Aging Research Center and Center for Alzheimer Research (YW, CQ), Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet-Stockholm University, 17165 Solna, Sweden
| |
Collapse
|
7
|
Adem MA, Decourt B, Sabbagh MN. Pharmacological Approaches Using Diabetic Drugs Repurposed for Alzheimer's Disease. Biomedicines 2024; 12:99. [PMID: 38255204 PMCID: PMC10813018 DOI: 10.3390/biomedicines12010099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) are chronic, progressive disorders affecting the elderly, which fosters global healthcare concern with the growing aging population. Both T2DM and AD have been linked with increasing age, advanced glycosylation end products, obesity, and insulin resistance. Insulin resistance in the periphery is significant in the development of T2DM and it has been posited that insulin resistance in the brain plays a key role in AD pathogenesis, earning AD the name "type 3 diabetes". These clinical and epidemiological links between AD and T2DM have become increasingly pronounced throughout the years, and serve as a means to investigate the effects of antidiabetic therapies in AD, such as metformin, intranasal insulin, incretins, DPP4 inhibitors, PPAR-γ agonists, SGLT2 inhibitors. The majority of these drugs have shown benefit in preclinical trials, and have shown some promising results in clinical trials, with the improvement of cognitive faculties in participants with mild cognitive impairment and AD. In this review, we have summarize the benefits, risks, and conflicting data that currently exist for diabetic drugs being repurposed for the treatment of AD.
Collapse
Affiliation(s)
- Muna A. Adem
- Department of Neurology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, 350 W. Thomas Rd., Phoenix, AZ 85013, USA
| | - Boris Decourt
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| | - Marwan N. Sabbagh
- Department of Neurology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, 350 W. Thomas Rd., Phoenix, AZ 85013, USA
| |
Collapse
|
8
|
Asano S, Ogawa A, Osada T, Oka S, Nakajima K, Oshima Y, Tanaka S, Kaga H, Tamura Y, Watada H, Kawamori R, Konishi S. Reduced gray matter volume in the default-mode network associated with insulin resistance. Cereb Cortex 2023; 33:11225-11234. [PMID: 37757477 DOI: 10.1093/cercor/bhad358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Insulin resistance may lead to structural and functional abnormalities of the human brain. However, the mechanism by which insulin resistance impairs the brain remains elusive. In this study, we used two large neuroimaging databases to investigate the brain regions where insulin resistance was associated with the gray matter volume and to examine the resting-state functional connectivity between these brain regions and each hypothalamic nucleus. Insulin resistance was associated with reduced gray matter volume in the regions of the default-mode and limbic networks in the cerebral cortex in older adults. Resting-state functional connectivity was prominent between these networks and the paraventricular nucleus of the hypothalamus, a hypothalamic interface connecting functionally with the cerebral cortex. Furthermore, we found a significant correlation in these networks between insulin resistance-related gray matter volume reduction and network paraventricular nucleus of the hypothalamus resting-state functional connectivity. These results suggest that insulin resistance-related gray matter volume reduction in the default-mode and limbic networks emerged through metabolic homeostasis mechanisms in the hypothalamus.
Collapse
Affiliation(s)
- Saki Asano
- Department of Neurophysiology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Akitoshi Ogawa
- Department of Neurophysiology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Takahiro Osada
- Department of Neurophysiology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Satoshi Oka
- Department of Neurophysiology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Koji Nakajima
- Department of Neurophysiology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Orthopaedic Surgery, The University of Tokyo School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yasushi Oshima
- Department of Orthopaedic Surgery, The University of Tokyo School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Sakae Tanaka
- Department of Orthopaedic Surgery, The University of Tokyo School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hideyoshi Kaga
- Department of Metabolism and Endocrinology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Sportology Center, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yoshifumi Tamura
- Department of Metabolism and Endocrinology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Sportology Center, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Hirotaka Watada
- Department of Metabolism and Endocrinology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Sportology Center, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Ryuzo Kawamori
- Department of Metabolism and Endocrinology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Sportology Center, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Seiki Konishi
- Department of Neurophysiology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Sportology Center, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Research Institute for Diseases of Old Age, Juntendo University School of Medicine , 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Advanced Research Institute for Health Science, Juntendo University School of Medicine , 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| |
Collapse
|
9
|
Wei B, Dong Q, Ma J, Zhang A. The association between triglyceride-glucose index and cognitive function in nondiabetic elderly: NHANES 2011-2014. Lipids Health Dis 2023; 22:188. [PMID: 37932783 PMCID: PMC10629120 DOI: 10.1186/s12944-023-01959-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND The relationship between Insulin resistance (IR) evaluated through homeostasis model assessment insulin resistance (HOMA-IR) and cognitive function is controversial among nondiabetic individuals. No study so far has reported the association between the IR evaluated through triglyceride glucose (TyG) index and cognitive function among nondiabetics. This study aims to assess this association among US nondiabetic older elderly. METHODS Data were obtained from the 2011-2014 National Health and Nutrition Examination Survey (NHANES). Low cognitive function was evaluated using the Consortium to Establish a Registry for Alzheimer's Disease Battery for immediate word list learning (CERAD-WL) and delayed recall (CERAD-DR) test, the Animal Fluency Test (AFT), and the Digit Symbol Substitution Test (DSST). Logistic regression analyses were conducted to compute the odds ratio (OR) and 95% confidential interval (CI) to examine the association between the TyG index (continuous and quartiles) and low cognitive function. RESULTS A total of 661 nondiabetic older adults were included with a mean age of 68.62 ± 6.49 years. Compared to the 1st quartile of the TyG index, participants in the TyG index 4th quartile were associated with low cognitive function evaluated through the CERAD test (CERAD-WL and CERAD-DR) [OR: 2.62; 95% CI (1.31, 5.23); P < 0.05]. Subgroup analyses showed that females (ORQ4 VS Q1: 3.07; 95% CI (1.04, 9.05); P < 0.05) and smokers (OR Q4 VS Q1: 2.70; 95% CI (1.01, 7.26); P < 0.05) categories were related with a higher risk of low cognitive function. CONCLUSIONS A high TyG index was strongly correlated with low cognitive function evaluated through the CERAD test (CERAD-WL and CERAD-DR) among US nondiabetic older women. The management of IR in women might be beneficial to primarily prevent low cognitive function among nondiabetic older elderly.
Collapse
Affiliation(s)
- Baojian Wei
- School of Nursing, Shandong First Medical University & Shandong Academy of Medical Sciences, No.619 Changcheng Road, Daiyue District, Taian, 271000, China
| | - Qianni Dong
- School of Nursing, Shandong First Medical University & Shandong Academy of Medical Sciences, No.619 Changcheng Road, Daiyue District, Taian, 271000, China
| | - Jinlong Ma
- School of Nursing, Yanbian University, Yanji, China
| | - Aihua Zhang
- School of Nursing, Shandong First Medical University & Shandong Academy of Medical Sciences, No.619 Changcheng Road, Daiyue District, Taian, 271000, China.
| |
Collapse
|
10
|
Li YS, Liu YL, Wang JJ, Haghbin N, Wang XH, Jiang WR, Qiu HN, Xia LF, Wu F, Lin CY, Li JB, Lin JN. Relationships Between Body Composition and Cognitive Impairment in Hospitalised Middle-Aged Type 2 Diabetic Patients. Diabetes Metab Syndr Obes 2023; 16:2389-2400. [PMID: 37581116 PMCID: PMC10423571 DOI: 10.2147/dmso.s418111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/29/2023] [Indexed: 08/16/2023] Open
Abstract
Objective The aim of this study was to elucidate the relationship between specific body composition and the risk of Cognitive Impairment (CI) in middle-aged Type 2 Diabetes Mellitus (T2DM) patients. Methods This cross-sectional study included 504 hospitalized patients with T2DM from the Department of Endocrinology and Metabolism of the Tianjin Union Medical Center. Subjects were grouped by sex, and cognitive status was assessed using the Montreal Cognitive Assessment (MoCA). The relationship between body composition and cognitive ability was investigated with the use of linear regression analysis. The association between body composition and CI risk was determined by logistic regression analysis. Results The prevalence of CI was 39.3% in middle-aged T2DM patients. After adjusting for age, education, marriage status, carotid atherosclerosis, cerebrovascular disease and hemoglobin, multiple linear regression analysis showed that lean mass index (LMI), body mass index (BMI) and appendicular skeletal muscle index (SMI) were significant predictors for the MoCA scores in men (p < 0.05). In addition, BMI (OR 0.913, 95% CI 0.840-0.992) and LMI (OR 0.820, 95% CI 0.682-0.916) were independent protective factors for CI in males. After adjusted for age, education, marriage status, dietary control of diabetes and cerebrovascular disease, visceral obesity (VO, OR 1.950, 95% CI 1.033-3.684) and abdominal obesity (AO, OR 2.537, 95% CI 1.191-5.403) were risk factors for CI in female patients. Conclusion The results suggest that there may be different mechanisms underlying the relationship of body compositions and cognitive performance between middle-aged male and female patients with T2DM. In addition, our finding of potential determinants of cognitive impairment may facilitate the development of intervention programs for middle-aged type 2 diabetic patients. Nevertheless, more large prospective studies looking at cognition and changes in body composition over time are needed in the future to further support their association.
Collapse
Affiliation(s)
- Yao-Shuang Li
- Tianjin Union Medical Center, Tianjin Medical University, Tianjin, People’s Republic of China
- Department of Endocrinology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, People’s Republic of China
| | - Yan-Lan Liu
- Department of Endocrinology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, People’s Republic of China
| | - Jun-Jia Wang
- Department of Endocrinology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, People’s Republic of China
- Tianjin Union Medical Center, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
| | - Nahal Haghbin
- School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Xiao-He Wang
- Institute of Non-Communicable Diseases Control and Prevention, Tianjin Center for Disease Control and Prevention, Tianjin, People’s Republic of China
| | - Wei-Ran Jiang
- Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, NY, USA
| | - Hui-Na Qiu
- Department of Endocrinology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, People’s Republic of China
| | - Long-Fei Xia
- Tianjin Union Medical Center, Tianjin Medical University, Tianjin, People’s Republic of China
- Department of Endocrinology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, People’s Republic of China
| | - Fan Wu
- Department of Endocrinology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, People’s Republic of China
- Tianjin Union Medical Center, School of Medicine, Nankai University, Tianjin, People’s Republic of China
| | - Chen-Ying Lin
- Tianjin Union Medical Center, Tianjin Medical University, Tianjin, People’s Republic of China
- Department of Endocrinology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, People’s Republic of China
| | - Jing-Bo Li
- Department of Endocrinology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, People’s Republic of China
| | - Jing-Na Lin
- Department of Endocrinology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, People’s Republic of China
| |
Collapse
|
11
|
Tian N, Fa W, Dong Y, Liu R, Liu C, Liu K, Mao M, Zhu M, Liang X, Wang N, Ma Y, Ngandu T, Launer LJ, Wang Y, Hou T, Du Y, Qiu C. Triglyceride-glucose index, Alzheimer's disease plasma biomarkers, and dementia in older adults: The MIND-China study. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12426. [PMID: 37101710 PMCID: PMC10123384 DOI: 10.1002/dad2.12426] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 02/04/2023] [Accepted: 03/12/2023] [Indexed: 04/28/2023]
Abstract
Introduction Population-based studies have rarely explored the associations of the triglyceride-glucose (TyG) index, a surrogate marker of insulin resistance, with dementia and plasma biomarkers for amyloid beta (Aβ) and neurodegeneration. Methods This population-based study included 5199 participants (age ≥ 65 years); of these, plasma Aβ, total tau, and neurofilament light chain (NfL) were measured in 1287 persons. Dementia and subtypes were diagnosed following the international criteria. TyG index was calculated as ln(fasting triglyceride(mg/dL) × fasting glucose[mg/dL]/2). Data were analyzed using logistic and general linear regression models. Results Dementia, Alzheimer's disease (AD), and vascular dementia (VaD) were diagnosed in 301, 195, and 95 individuals, respectively. A high TyG index was significantly associated with increased likelihoods of dementia and AD; the significant association with dementia remained among participants without cardiovascular disease or diabetes. In the biomarker subsample, a high TyG index was correlated with elevated plasma Aβ, but not with total tau or NfL. Discussion High TyG index is associated with dementia, possibly via Aβ pathology.
Collapse
Affiliation(s)
- Na Tian
- Department of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanP.R. China
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanP.R. China
| | - Wenxin Fa
- Department of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanP.R. China
| | - Yi Dong
- Department of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanP.R. China
| | - Rui Liu
- Department of NeurologyShandong Provincial HospitalShandong UniversityJinanP.R. China
| | - Cuicui Liu
- Department of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanP.R. China
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanP.R. China
| | - Keke Liu
- Department of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanP.R. China
| | - Ming Mao
- Department of NeurologyShandong Provincial HospitalShandong UniversityJinanP.R. China
| | - Min Zhu
- Department of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanP.R. China
| | - Xiaoyan Liang
- Department of NeurologyShandong Provincial HospitalShandong UniversityJinanP.R. China
| | - Nan Wang
- Department of NeurologyShandong Provincial HospitalShandong UniversityJinanP.R. China
| | - Yixun Ma
- Department of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanP.R. China
| | - Tiia Ngandu
- Division of Clinical Geriatrics and Center for Alzheimer ResearchDepartment of NeurobiologyCare Sciences and Society (NVS), Karolinska InstitutetStockholmSweden
- Department of Public Health and WelfareFinnish Institute for Health and WelfareHelsinkiFinland
| | - Lenore J. Launer
- Intramural Research Program, Laboratory of Epidemiology and Population SciencesNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - Yongxiang Wang
- Department of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanP.R. China
- Aging Research Center and Center for Alzheimer ResearchDepartment of NVSKarolinska Institutet‐Stockholm UniversityStockholmSweden
| | - Tingting Hou
- Department of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanP.R. China
| | - Yifeng Du
- Department of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanP.R. China
- Shandong Provincial Clinical Research Center for Neurological DiseasesJinanP.R. China
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanP.R. China
- Department of NeurologyShandong Provincial HospitalShandong UniversityJinanP.R. China
| | - Chengxuan Qiu
- Department of NeurologyShandong Provincial Hospital affiliated to Shandong First Medical UniversityJinanP.R. China
- Aging Research Center and Center for Alzheimer ResearchDepartment of NVSKarolinska Institutet‐Stockholm UniversityStockholmSweden
| |
Collapse
|
12
|
Mazza E, Calesella F, Paolini M, di Pasquasio C, Poletti S, Lorenzi C, Falini A, Zanardi R, Colombo C, Benedetti F. Insulin resistance disrupts white matter microstructure and amplitude of functional spontaneous activity in bipolar disorder. Bipolar Disord 2023; 25:32-42. [PMID: 36377438 DOI: 10.1111/bdi.13270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Bipolar disorder (BD) is linked to several structural and functional brain alterations. In addition, BD patients have a three-fold increased risk of developing insulin resistance, which is associated with neural changes and poorer BD outcomes. Therefore, we investigated the effects of insulin and two derived measures (insulin resistance and sensitivity) on white matter (WM) microstructure, resting-state (rs) functional connectivity (FC), and fractional amplitude of low-frequency fluctuation (fALFF). METHODS BD patients (n = 92) underwent DTI acquisition, and a subsample (n = 22) underwent rs-fMRI. Blood samples were collected to determine insulin and glucose levels. The Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) and quantitative insulin sensitivity check index (QUICKI) were computed. DTI data were analyzed via tract-based spatial statistics and threshold-free cluster enhancement. From rs-fMRI data, both ROI-to-ROI FC matrices and fALFF maps were extracted. RESULTS Insulin showed a widespread negative association with fractional anisotropy (FA) and a positive effect on radial diffusivity (RD) and mean diffusivity (MD). HOMA-IR exerted a significant effect on RD in the right superior longitudinal fasciculus, whereas QUICKI was positively associated with FA and negatively with RD and MD in the left superior longitudinal fasciculus, left anterior corona radiata, and forceps minor. fALFF was negatively modulated by insulin and HOMA-IR and positively associated with QUICKI in the precuneus. No significant results were found in the ROI-to-ROI analysis. CONCLUSION Our findings suggest that WM microstructure and functional alterations might underlie the effect of IR on BD pathophysiology, even if the causal mechanisms need to be further investigated.
Collapse
Affiliation(s)
- Elena Mazza
- Vita-Salute San Raffaele University, Milan, Italy.,Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute IRCCS Ospedale San Raffaele, Milan, Italy
| | - Federico Calesella
- Vita-Salute San Raffaele University, Milan, Italy.,Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute IRCCS Ospedale San Raffaele, Milan, Italy
| | - Marco Paolini
- Vita-Salute San Raffaele University, Milan, Italy.,Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Sara Poletti
- Vita-Salute San Raffaele University, Milan, Italy.,Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute IRCCS Ospedale San Raffaele, Milan, Italy
| | - Cristina Lorenzi
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute IRCCS Ospedale San Raffaele, Milan, Italy
| | - Andrea Falini
- Vita-Salute San Raffaele University, Milan, Italy.,C.E.R.M.A.C. (Centro di Eccellenza Risonanza Magnetica ad Alto Campo), University Vita-Salute San Raffaele, Milan, Italy
| | - Raffaella Zanardi
- Vita-Salute San Raffaele University, Milan, Italy.,Mood Disorders Unit, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Cristina Colombo
- Vita-Salute San Raffaele University, Milan, Italy.,Mood Disorders Unit, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Francesco Benedetti
- Vita-Salute San Raffaele University, Milan, Italy.,Psychiatry and Clinical Psychobiology, Division of Neuroscience, Scientific Institute IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
13
|
Cui Y, Tang TY, Lu CQ, Ju S. Insulin Resistance and Cognitive Impairment: Evidence From Neuroimaging. J Magn Reson Imaging 2022; 56:1621-1649. [PMID: 35852470 DOI: 10.1002/jmri.28358] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/02/2022] [Accepted: 07/05/2022] [Indexed: 01/04/2023] Open
Abstract
Insulin is a peptide well known for its role in regulating glucose metabolism in peripheral tissues. Emerging evidence from human and animal studies indicate the multifactorial role of insulin in the brain, such as neuronal and glial metabolism, glucose regulation, and cognitive processes. Insulin resistance (IR), defined as reduced sensitivity to the action of insulin, has been consistently proposed as an important risk factor for developing neurodegeneration and cognitive impairment. Although the exact mechanism of IR-related cognitive impairment still awaits further elucidation, neuroimaging offers a versatile set of novel contrasts to reveal the subtle cerebral abnormalities in IR. These imaging contrasts, including but not limited to brain volume, white matter (WM) microstructure, neural function and brain metabolism, are expected to unravel the nature of the link between IR, cognitive decline, and brain abnormalities, and their changes over time. This review summarizes the current neuroimaging studies with multiparametric techniques, focusing on the cerebral abnormalities related to IR and therapeutic effects of IR-targeting treatments. According to the results, brain regions associated with IR pathophysiology include the medial temporal lobe, hippocampus, prefrontal lobe, cingulate cortex, precuneus, occipital lobe, and the WM tracts across the globe. Of these, alterations in the temporal lobe are highly reproducible across different imaging modalities. These structures have been known to be vulnerable to Alzheimer's disease (AD) pathology and are critical in cognitive processes such as memory and executive functioning. Comparing to asymptomatic subjects, results are more mixed in patients with metabolic disorders such as type 2 diabetes and obesity, which might be attributed to a multifactorial mechanism. Taken together, neuroimaging, especially MRI, is beneficial to reveal early abnormalities in cerebral structure and function in insulin-resistant brain, providing important evidence to unravel the underlying neuronal substrate that reflects the cognitive decline in IR. EVIDENCE LEVEL: 5 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Ying Cui
- Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Tian-Yu Tang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Chun-Qiang Lu
- Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Shenghong Ju
- Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
14
|
Low molecular weight fucoidan alleviates cerebrovascular damage by promoting angiogenesis in type 2 diabetes mice. Int J Biol Macromol 2022; 217:345-355. [PMID: 35841956 DOI: 10.1016/j.ijbiomac.2022.07.053] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 12/11/2022]
Abstract
Diabetes leading to brain glucose metabolism disorders and cerebrovascular complications. Fucoidan is a kind of sulfated polysaccharides which found in brown algae, has multiply bioactivities and considered to be a promising therapeutic agent. Despite the increasing amount of evidence suggesting the diabetes protective role of fucoidans, the effect of fucoidan on brain abnormalities in type 2 diabetes mellitus patients remains unclear. In this study a low molecular weight fucoidan (LMWF) was obtained from Saccharina japonica and its effect on the cerebrovascular damage in db/db mice was investigated. Results were shown after LMWF treatment, the degree of cerebrovascular damage, the number of apoptotic neuronal cells and the inflammation were all decreased in db/db mice. Moreover, LMWF could up-regulates CD34 and VEGFA expression in db/db mice brain, and the subintestinal vessel angiogenesis in zebrafish was also promoted by LMWF. Moreover, the lumen formation of HUVEC endothelial cells was rescued by LMWF which was destroyed in high glucose treated endothelial cells. Further study found, LMWF alleviates vascular injury by up-regulating the expression level of phosphorylated PI3K and phosphorylated AKT. Our study indicates that LMWF has the potential to develop a cerebrovascular protection agent for type 2 diabetes patients.
Collapse
|
15
|
Javaid MA, Selim M, Ortega-Gutierrez S, Lattanzi S, Zargar S, Alaouieh DA, Hong E, Divani AA. Potential application of intranasal insulin delivery for treatment of intracerebral hemorrhage: A review of the literature. J Stroke Cerebrovasc Dis 2022; 31:106489. [PMID: 35489182 DOI: 10.1016/j.jstrokecerebrovasdis.2022.106489] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 03/04/2022] [Accepted: 04/03/2022] [Indexed: 12/01/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating subtype of stroke associated with high morbidity and mortality that is considered a medical emergency, mainly managed with adequate blood pressure control and creating a favorable hemostatic condition. However, to date, none of the randomized clinical trials have led to an effective treatment for ICH. It is vital to better understand the mechanisms underlying brain injury to effectively decrease ICH-associated morbidity and mortality. It is well known that initial hematoma formation and its expansion have detrimental consequences. The literature has recently focused on other pathological processes, including oxidative stress, neuroinflammation, blood-brain barrier disruption, edema formation, and neurotoxicity, that constitute secondary brain injury. Since conventional management has failed to improve clinical outcomes significantly, various neuroprotective therapies are tested in preclinical and clinical settings. Unlike intravenous administration, intranasal insulin can reach a higher concentration in the cerebrospinal fluid without causing systemic side effects. Intranasal insulin delivery has been introduced as a novel neuroprotective agent for certain neurological diseases, including ischemic stroke, subarachnoid hemorrhage, and traumatic brain injury. Since there is an overlap of mechanisms causing neuroinflammation in these neurological diseases and ICH, we believe that preclinical studies testing the role of intranasal insulin therapy in ICH are warranted.
Collapse
Affiliation(s)
| | - Magdy Selim
- Stroke Division, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Simona Lattanzi
- Neurological Clinic, Department of Experimental and Clinical Medicine, Marche Polytechnic University, Ancona, Italy
| | - Shima Zargar
- Department of Neurology, University of New Mexico, Albuquerque, NM, USA
| | | | - Emily Hong
- School of Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Afshin A Divani
- Department of Neurology, University of New Mexico, Albuquerque, NM, USA.
| |
Collapse
|
16
|
Xue P, Tang X, Tan X, Benedict C. Self-reported regular daytime napping is associated with indicators of poor type 2 diabetes control: A cohort study. Diabetes Obes Metab 2022; 24:737-741. [PMID: 34882924 DOI: 10.1111/dom.14619] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/30/2021] [Accepted: 12/06/2021] [Indexed: 02/05/2023]
Affiliation(s)
- Pei Xue
- Department of Surgical Sciences, Sleep Science Laboratory (BMC), Uppsala University, Uppsala, Sweden
- Sleep Medicine Center, Department of Respiratory and Critical Care Medicine, Mental Health Center, Translational Neuroscience Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangdong Tang
- Sleep Medicine Center, Department of Respiratory and Critical Care Medicine, Mental Health Center, Translational Neuroscience Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiao Tan
- Department of Surgical Sciences, Sleep Science Laboratory (BMC), Uppsala University, Uppsala, Sweden
| | - Christian Benedict
- Department of Surgical Sciences, Sleep Science Laboratory (BMC), Uppsala University, Uppsala, Sweden
| |
Collapse
|
17
|
Woodfield A, Porter T, Gilani I, Noordin S, Li QX, Collins S, Martins RN, Maruff P, Masters CL, Rowe CC, Villemagne VL, Dore V, Newsholme P, Laws SM, Verdile G. Insulin resistance, cognition and Alzheimer's disease biomarkers: Evidence that CSF Aβ42 moderates the association between insulin resistance and increased CSF tau levels. Neurobiol Aging 2022; 114:38-48. [DOI: 10.1016/j.neurobiolaging.2022.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/09/2022] [Accepted: 03/07/2022] [Indexed: 12/16/2022]
|
18
|
Mucellini AB, Miguel PM, Dalle Molle R, Rodrigues DM, Machado TD, Reis RS, Toazza R, Salum GA, Bortoluzzi A, Franco AR, Buchweitz A, Barth B, Agranonik M, Nassim M, Meaney MJ, Manfro GG, Silveira PP. Diminished insulin sensitivity is associated with altered brain activation to food cues and with risk for obesity - Implications for individuals born small for gestational age. Appetite 2021; 169:105799. [PMID: 34767841 DOI: 10.1016/j.appet.2021.105799] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 10/14/2021] [Accepted: 11/05/2021] [Indexed: 11/02/2022]
Abstract
While classically linked to memory, the hippocampus is also a feeding behavior modulator due to its multiple interconnected pathways with other brain regions and expression of receptors for metabolic hormones. Here we tested whether variations in insulin sensitivity would be correlated with differential brain activation following exposure to palatable food cues, as well as with variations in implicit food memory in a cohort of healthy adolescents, some of whom were born small for gestational age (SGA). Homeostatic Model Assessment of Insulin Resistance (HOMA-IR) was positively correlated with activation in the cuneus, and negatively correlated with activation in the middle frontal lobe, superior frontal gyrus and precuneus when presented with palatable food images versus non-food images in healthy adolescents. Additionally, HOMA-IR and insulinemia were higher in participants with impaired food memory. SGA individuals had higher snack caloric density and greater chance for impaired food memory. There was also an interaction between the HOMA-IR and birth weight ratio influencing external eating behavior. We suggest that diminished insulin sensitivity correlates with activation in visual attention areas and inactivation in inhibitory control areas in healthy adolescents. Insulin resistance also associated with less consistency in implicit memory for a consumed meal, which may suggest lower ability to establish a dietary pattern, and can contribute to obesity. Differences in feeding behavior in SGA individuals were associated with insulin sensitivity and hippocampal alterations, suggesting that cognition and hormonal regulation are important components involved in their food intake modifications throughout life.
Collapse
Affiliation(s)
- Amanda B Mucellini
- Graduate Program in Psychiatry and Behavioral Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Patrícia M Miguel
- Graduate Program in Neuroscience, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Roberta Dalle Molle
- Graduate Program in Child and Adolescent Health, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Danitsa M Rodrigues
- Graduate Program in Neuroscience, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Tania D Machado
- Graduate Program in Child and Adolescent Health, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Roberta S Reis
- Graduate Program in Child and Adolescent Health, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Rudinéia Toazza
- Graduate Program in Neuroscience, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Giovanni A Salum
- Graduate Program in Psychiatry and Behavioral Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Andressa Bortoluzzi
- Graduate Program in Neuroscience, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Alexandre R Franco
- Brain Institute of Rio Grande do Sul, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Augusto Buchweitz
- Brain Institute of Rio Grande do Sul, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Barbara Barth
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada; Integrated Program in Neurosciences, McGill University, Montreal, QC, Canada
| | - Marilyn Agranonik
- Fundação de Economia e Estatística Siegfried Emanuel Heuser, Porto Alegre, Brazil
| | - Marouane Nassim
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada
| | - Michael J Meaney
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Translational Neuroscience Programme, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Gisele G Manfro
- Graduate Program in Psychiatry and Behavioral Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Graduate Program in Neuroscience, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Patrícia P Silveira
- Graduate Program in Neuroscience, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
19
|
Ennis GE, Kohli A, Jonaitis EM, Betthauser TJ, Oh JM, Taylor CE, Chin N, Koscik RL, Christian BT, Asthana S, Johnson SC, Bendlin BB. The relationship of glucose-stimulated insulin secretion to cerebral glucose metabolism and cognition in healthy middle-aged and older adults. Neurobiol Aging 2021; 105:174-185. [PMID: 34091125 PMCID: PMC8338794 DOI: 10.1016/j.neurobiolaging.2021.04.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 11/04/2020] [Accepted: 04/27/2021] [Indexed: 01/04/2023]
Abstract
Insulin resistance (IR) has been related to reduced cerebral glucose metabolism in regions identified as hypometabolic in Alzheimer's clinical syndrome. Insulin secretion (IS) has been less studied than IR despite findings that decreased IS is an early indicator of future type 2 diabetes and a potential predictor of Alzheimer's clinical syndrome. We investigated whether higher IR and lower IS would be associated with greater age-related reductions in regional cerebral glucose metabolism and worse cognitive performance. Two-hour oral glucose tolerance testing and 18F-fluorodeoxyglucose positron emission tomography were performed on 1-2 occasions on a sample of healthy middle-aged and older adults from the Wisconsin Alzheimer's Disease Research Center. Neuropsychological tests were completed during Alzheimer's Disease Research Center Clinical Core visits. Pattern of findings suggested that lower (not higher) IS was related to higher regional cerebral glucose metabolism in middle aged but not older adults, and lower (not higher) IS was also related to better immediate recall. In the context of healthy insulin sensitivity, lower IS may be beneficial to brain health.
Collapse
Affiliation(s)
- Gilda E Ennis
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | - Akshay Kohli
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Erin M Jonaitis
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Tobey J Betthauser
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jennifer M Oh
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Chase E Taylor
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Nathaniel Chin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Rebecca L Koscik
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Bradley T Christian
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison, Madison, WI, USA; Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Geriatric Research Education and Clinical Center, William S. Middleton Hospital Department of Veterans Affairs, Madison, WI, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Geriatric Research Education and Clinical Center, William S. Middleton Hospital Department of Veterans Affairs, Madison, WI, USA
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Geriatric Research Education and Clinical Center, William S. Middleton Hospital Department of Veterans Affairs, Madison, WI, USA
| |
Collapse
|
20
|
Diabetes and Alzheimer's Disease: Might Mitochondrial Dysfunction Help Deciphering the Common Path? Antioxidants (Basel) 2021; 10:antiox10081257. [PMID: 34439505 PMCID: PMC8389322 DOI: 10.3390/antiox10081257] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 02/06/2023] Open
Abstract
A growing number of clinical and epidemiological studies support the hypothesis of a tight correlation between type 2 diabetes mellitus (T2DM) and the development risk of Alzheimer's disease (AD). Indeed, the proposed definition of Alzheimer's disease as type 3 diabetes (T3D) underlines the key role played by deranged insulin signaling to accumulation of aggregated amyloid beta (Aβ) peptides in the senile plaques of the brain. Metabolic disturbances such as hyperglycemia, peripheral hyperinsulinemia, dysregulated lipid metabolism, and chronic inflammation associated with T2DM are responsible for an inefficient transport of insulin to the brain, producing a neuronal insulin resistance that triggers an enhanced production and deposition of Aβ and concomitantly contributes to impairment in the micro-tubule-associated protein Tau, leading to neural degeneration and cognitive decline. Furthermore, the reduced antioxidant capacity observed in T2DM patients, together with the impairment of cerebral glucose metabolism and the decreased performance of mitochondrial activity, suggests the existence of a relationship between oxidative damage, mitochondrial impairment, and cognitive dysfunction that could further reinforce the common pathophysiology of T2DM and AD. In this review, we discuss the molecular mechanisms by which insulin-signaling dysregulation in T2DM can contribute to the pathogenesis and progression of AD, deepening the analysis of complex mechanisms involved in reactive oxygen species (ROS) production under oxidative stress and their possible influence in AD and T2DM. In addition, the role of current therapies as tools for prevention or treatment of damage induced by oxidative stress in T2DM and AD will be debated.
Collapse
|
21
|
Saunders AM, Burns DK, Gottschalk WK. Reassessment of Pioglitazone for Alzheimer's Disease. Front Neurosci 2021; 15:666958. [PMID: 34220427 PMCID: PMC8243371 DOI: 10.3389/fnins.2021.666958] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/18/2021] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease is a quintessential 'unmet medical need', accounting for ∼65% of progressive cognitive impairment among the elderly, and 700,000 deaths in the United States in 2020. In 2019, the cost of caring for Alzheimer's sufferers was $244B, not including the emotional and physical toll on caregivers. In spite of this dismal reality, no treatments are available that reduce the risk of developing AD or that offer prolonged mitiagation of its most devestating symptoms. This review summarizes key aspects of the biology and genetics of Alzheimer's disease, and we describe how pioglitazone improves many of the patholophysiological determinants of AD. We also summarize the results of pre-clinical experiments, longitudinal observational studies, and clinical trials. The results of animal testing suggest that pioglitazone can be corrective as well as protective, and that its efficacy is enhanced in a time- and dose-dependent manner, but the dose-effect relations are not monotonic or sigmoid. Longitudinal cohort studies suggests that it delays the onset of dementia in individuals with pre-existing type 2 diabetes mellitus, which small scale, unblinded pilot studies seem to confirm. However, the results of placebo-controlled, blinded clinical trials have not borne this out, and we discuss possible explanations for these discrepancies.
Collapse
Affiliation(s)
- Ann M. Saunders
- Zinfandel Pharmaceuticals, Inc., Chapel Hill, NC, United States
| | - Daniel K. Burns
- Zinfandel Pharmaceuticals, Inc., Chapel Hill, NC, United States
| | | |
Collapse
|
22
|
McWhinney S, Kolenic M, Franke K, Fialova M, Knytl P, Matejka M, Spaniel F, Hajek T. Obesity as a Risk Factor for Accelerated Brain Ageing in First-Episode Psychosis-A Longitudinal Study. Schizophr Bull 2021; 47:1772-1781. [PMID: 34080013 PMCID: PMC8530396 DOI: 10.1093/schbul/sbab064] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Obesity is highly prevalent in schizophrenia, with implications for psychiatric prognosis, possibly through links between obesity and brain structure. In this longitudinal study in first episode of psychosis (FEP), we used machine learning and structural magnetic resonance imaging (MRI) to study the impact of psychotic illness and obesity on brain ageing/neuroprogression shortly after illness onset. METHODS We acquired 2 prospective MRI scans on average 1.61 years apart in 183 FEP and 155 control individuals. We used a machine learning model trained on an independent sample of 504 controls to estimate the individual brain ages of study participants and calculated BrainAGE by subtracting chronological from the estimated brain age. RESULTS Individuals with FEP had a higher initial BrainAGE than controls (3.39 ± 6.36 vs 1.72 ± 5.56 years; β = 1.68, t(336) = 2.59, P = .01), but similar annual rates of brain ageing over time (1.28 ± 2.40 vs 1.07±1.74 estimated years/actual year; t(333) = 0.93, P = .18). Across both cohorts, greater baseline body mass index (BMI) predicted faster brain ageing (β = 0.08, t(333) = 2.59, P = .01). For each additional BMI point, the brain aged by an additional month per year. Worsening of functioning over time (Global Assessment of Functioning; β = -0.04, t(164) = -2.48, P = .01) and increases especially in negative symptoms on the Positive and Negative Syndrome Scale (β = 0.11, t(175) = 3.11, P = .002) were associated with faster brain ageing in FEP. CONCLUSIONS Brain alterations in psychosis are manifest already during the first episode and over time get worse in those with worsening clinical outcomes or higher baseline BMI. As baseline BMI predicted faster brain ageing, obesity may represent a modifiable risk factor in FEP that is linked with psychiatric outcomes via effects on brain structure.
Collapse
Affiliation(s)
- Sean McWhinney
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Marian Kolenic
- National Institute of Mental Health, Klecany, Czech Republic,Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Katja Franke
- Structural Brain Mapping Group, Department of Neurology, Jena University Hospital, Jena, Germany
| | - Marketa Fialova
- National Institute of Mental Health, Klecany, Czech Republic
| | - Pavel Knytl
- National Institute of Mental Health, Klecany, Czech Republic
| | - Martin Matejka
- National Institute of Mental Health, Klecany, Czech Republic,Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Filip Spaniel
- National Institute of Mental Health, Klecany, Czech Republic,Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Tomas Hajek
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada,National Institute of Mental Health, Klecany, Czech Republic,To whom correspondence should be addressed; Department of Psychiatry, Dalhousie University, QEII HSC, A. J. Lane Building, Room 3093, 5909 Veteran’s Memorial Lane, Halifax, Nova Scotia B3H 2E2, Canada; tel: (902) 473-8299, fax: (902) 473-1583, e-mail:
| |
Collapse
|
23
|
Chen Q, Cao T, Li N, Zeng C, Zhang S, Wu X, Zhang B, Cai H. Repurposing of Anti-Diabetic Agents as a New Opportunity to Alleviate Cognitive Impairment in Neurodegenerative and Neuropsychiatric Disorders. Front Pharmacol 2021; 12:667874. [PMID: 34108878 PMCID: PMC8182376 DOI: 10.3389/fphar.2021.667874] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/07/2021] [Indexed: 12/16/2022] Open
Abstract
Cognitive impairment is a shared abnormality between type 2 diabetes mellitus (T2DM) and many neurodegenerative and neuropsychiatric disorders, such as Alzheimer’s disease (AD) and schizophrenia. Emerging evidence suggests that brain insulin resistance plays a significant role in cognitive deficits, which provides the possibility of anti-diabetic agents repositioning to alleviate cognitive deficits. Both preclinical and clinical studies have evaluated the potential cognitive enhancement effects of anti-diabetic agents targeting the insulin pathway. Repurposing of anti-diabetic agents is considered to be promising for cognitive deficits prevention or control in these neurodegenerative and neuropsychiatric disorders. This article reviewed the possible relationship between brain insulin resistance and cognitive deficits. In addition, promising therapeutic interventions, especially current advances in anti-diabetic agents targeting the insulin pathway to alleviate cognitive impairment in AD and schizophrenia were also summarized.
Collapse
Affiliation(s)
- Qian Chen
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Ting Cao
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - NaNa Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Cuirong Zeng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Shuangyang Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Xiangxin Wu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Bikui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Hualin Cai
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| |
Collapse
|
24
|
Abstract
The intranasal (IN) route enables the delivery of insulin to the central nervous system in the relative absence of systemic uptake and related peripheral side effects. Intranasally administered insulin is assumed to travel along olfactory and adjacent pathways and has been shown to rapidly accumulate in cerebrospinal fluid, indicating efficient transport to the brain. Two decades of studies in healthy humans and patients have demonstrated that IN insulin exerts functional effects on metabolism, such as reductions in food intake and body weight and improvements of glucose homeostasis, as well as cognition, ie, enhancements of memory performance both in healthy individuals and patients with mild cognitive impairment or Alzheimer's disease; these studies moreover indicate a favourable safety profile of the acute and repeated use of IN insulin. Emerging findings suggest that IN insulin also modulates neuroendocrine activity, sleep-related mechanisms, sensory perception and mood. Some, but not all studies point to sex differences in the response to IN insulin that need to be further investigated along with the impact of age. "Brain insulin resistance" is an evolving concept that posits impairments in central nervous insulin signalling as a pathophysiological factor in metabolic and cognitive disorders such as obesity, type 2 diabetes and Alzheimer's disease, and, notably, a target of interventions that rely on IN insulin. Still, the negative outcomes of longer-term IN insulin trials in individuals with obesity or Alzheimer's disease highlight the need for conceptual as well as methodological advances to translate the promising results of proof-of-concept experiments and pilot clinical trials into the successful clinical application of IN insulin.
Collapse
Affiliation(s)
- Manfred Hallschmid
- Institute of Medical Psychology and Behavioural Neurobiology, University of Tübingen, Tübingen, Germany
- German Centre for Diabetes Research (DZD), Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
| |
Collapse
|
25
|
Lu R, Aziz NA, Diers K, Stöcker T, Reuter M, Breteler MMB. Insulin resistance accounts for metabolic syndrome-related alterations in brain structure. Hum Brain Mapp 2021; 42:2434-2444. [PMID: 33769661 PMCID: PMC8090787 DOI: 10.1002/hbm.25377] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 01/22/2021] [Accepted: 01/28/2021] [Indexed: 12/26/2022] Open
Abstract
Metabolic syndrome (MetS) is a major public health burden worldwide and associated with brain abnormalities. Although insulin resistance is considered a pivotal feature of MetS, its role in the pathogenesis of MetS‐related brain alterations in the general population is unclear. Therefore, in 973 participants (mean age 52.5 years) of the population‐based Rhineland Study, we assessed brain morphology in relation to MetS and insulin resistance, and evaluated to what extent the pattern of structural brain changes seen in MetS overlap with those associated with insulin resistance. Cortical reconstruction and volumetric segmentation were obtained from high‐resolution brain images at 3 Tesla using FreeSurfer. The relations between metabolic measures and brain structure were assessed through (generalized) linear models. Both MetS and insulin resistance were associated with smaller cortical gray matter volume and thickness, but not with white matter or subcortical gray matter volume. Age‐ and sex‐adjusted vertex‐based brain morphometry demonstrated that MetS and insulin resistance were related to cortical thinning in a similar spatial pattern. Importantly, no independent effect of MetS on cortical gray matter was observed beyond the effect of insulin resistance. Our findings suggest that addressing insulin resistance is critical in the prevention of MetS‐related brain changes in later life.
Collapse
Affiliation(s)
- Ran Lu
- Population Health Sciences, German Center for Neurodegenerative diseases (DZNE), Bonn, Germany
| | - N Ahmad Aziz
- Population Health Sciences, German Center for Neurodegenerative diseases (DZNE), Bonn, Germany.,Department of Neurology, Faculty of Medicine, University of Bonn, Bonn, Germany
| | - Kersten Diers
- Image Analysis, German Center for Neurodegenerative diseases (DZNE), Bonn, Germany
| | - Tony Stöcker
- MR Physics, German Center for Neurodegenerative diseases (DZNE), Bonn, Germany.,Department of Physics and Astronomy, University of Bonn, Bonn, Germany
| | - Martin Reuter
- Image Analysis, German Center for Neurodegenerative diseases (DZNE), Bonn, Germany.,A.A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA.,Department of Radiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Monique M B Breteler
- Population Health Sciences, German Center for Neurodegenerative diseases (DZNE), Bonn, Germany.,Institute for Medical Biometry, Informatics and Epidemiology (IMBIE), Faculty of Medicine, University of Bonn, Bonn, Germany
| |
Collapse
|
26
|
Raiko JRH, Tuulari JJ, Saari T, Parkkola R, Savisto N, Nuutila P, Virtanen K. Associations Between Brain Gray Matter Volumes and Adipose Tissue Metabolism in Healthy Adults. Obesity (Silver Spring) 2021; 29:543-549. [PMID: 33528921 DOI: 10.1002/oby.23094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/12/2020] [Accepted: 11/12/2020] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Gray matter (GM) volume in different brain loci has been shown to vary in obesity and diabetes, and elevated fasting plasma nonesterified fatty acid (NEFA) levels have been suggested as one potential mechanism. The hypothesis presented in this study is that brown adipose tissue (BAT) activity may correlate with GM volume in areas negatively associated with obesity and diabetes. METHODS A total of 36 healthy patients (M/F: 12/24, age 39.7 ± 9.4 years, BMI 27.5 ± 5.6 kg/m2 ) were imaged with positron emission tomography using fatty acid analog [18 F]FTHA to measure NEFA uptake and with [15 O]H2 O to measure perfusion during cold exposure, at room temperature during fasting, or during a postprandial state. A 2-hour hyperinsulinemic euglycemic clamp was performed to measure whole-body insulin sensitivity (M value, mean 7.6 ± 3.9 mg/kg/min). T1-weighted magnetic resonance imaging at 1.5 T was performed on all patients. RESULTS BAT NEFA uptake was associated directly with GM volume in anterior cerebellum and occipital lobe (P ≤ 0.04) when adjusted for age, gender, and intra-abdominal fat volume and with anterior cerebellum, limbic lobe, and temporal lobe GM volumes when adjusted for M value. CONCLUSIONS BAT NEFA metabolism may participate in protection from cognitive degeneration associated with cardiometabolic risk factors, such as central obesity and insulin resistance. Potential causal relationships between BAT activity and GM volumes remain to be examined.
Collapse
Affiliation(s)
- Juho R H Raiko
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Jetro J Tuulari
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Turku, Finland
- Department of Endocrinology, Turku University Hospital, Turku, Finland
- Department of Psychiatry, University of Turku and Turku University Hospital, Turku, Finland
- Turku Collegium for Science and Medicine, University of Turku, Turku, Finland
| | - Teemu Saari
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Riitta Parkkola
- Department of Radiology, Turku University Hospital, Turku, Finland
| | - Nina Savisto
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Pirjo Nuutila
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
- Department of Endocrinology, Turku University Hospital, Turku, Finland
| | - Kirsi Virtanen
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| |
Collapse
|
27
|
Abstract
Brain insulin signaling contributes to memory function and might be a viable target in the prevention and treatment of memory impairments including Alzheimer's disease. This short narrative review explores the potential of central nervous system (CNS) insulin administration via the intranasal pathway to improve memory performance in health and disease, with a focus on the most recent results. Proof-of-concept studies and (pilot) clinical trials in individuals with mild cognitive impairment or Alzheimer's disease indicate that acute and prolonged intranasal insulin administration enhances memory performance, and suggest that brain insulin resistance is a pathophysiological factor in Alzheimer's disease with or without concomitant metabolic dysfunction. Intranasally administered insulin is assumed to trigger improvements in synaptic plasticity and regional glucose uptake as well as alleviations of Alzheimer's disease neuropathology; additional contributions of changes in hypothalamus-pituitary-adrenocortical axis activity and sleep-related mechanisms are discussed. While intranasal insulin delivery has been conclusively demonstrated to be effective and safe, the recent outcomes of large-scale clinical studies underline the need for further investigations, which might also yield new insights into sex differences in the response to intranasal insulin and contribute to the optimization of delivery devices to grasp the full potential of intranasal insulin for Alzheimer's disease.
Collapse
Affiliation(s)
- Manfred Hallschmid
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Otfried-Müller-Str. 25, 72076, Tübingen, Germany.
- German Center for Diabetes Research (DZD), Tübingen, Germany.
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.
| |
Collapse
|
28
|
Morris JK, John CS, Green ZD, Wilkins HM, Wang X, Kamat A, Swerdlow RS, Vidoni ED, Petersen ME, O’Bryant SE, Honea RA, Burns JM. Characterization of the Meal-Stimulated Incretin Response and Relationship With Structural Brain Outcomes in Aging and Alzheimer's Disease. Front Neurosci 2020; 14:608862. [PMID: 33328877 PMCID: PMC7734152 DOI: 10.3389/fnins.2020.608862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/02/2020] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Individuals with Alzheimer's Disease (AD) are often characterized by systemic markers of insulin resistance; however, the broader effects of AD on other relevant metabolic hormones, such as incretins that affect insulin secretion and food intake, remains less clear. METHODS Here, we leveraged a physiologically relevant meal tolerance test to assess diagnostic differences in these metabolic responses in cognitively healthy older adults (CH; n = 32) and AD (n = 23) participants. All individuals also underwent a comprehensive clinical examination, cognitive evaluation, and structural magnetic resonance imaging. RESULTS The meal-stimulated response of glucose, insulin, and peptide tyrosine tyrosine (PYY) was significantly greater in individuals with AD as compared to CH. Voxel-based morphometry revealed negative relationships between brain volume and the meal-stimulated response of insulin, C-Peptide, and glucose-dependent insulinotropic polypeptide (GIP) in primarily parietal brain regions. CONCLUSION Our findings are consistent with prior work that shows differences in metabolic regulation in AD and relationships with cognition and brain structure.
Collapse
Affiliation(s)
- Jill K. Morris
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, United States
| | - Casey S. John
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, United States
| | - Zachary D. Green
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, United States
| | - Heather M. Wilkins
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, United States
| | - Xiaowan Wang
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, United States
| | - Ashwini Kamat
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, United States
| | - Russell S. Swerdlow
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, United States
| | - Eric D. Vidoni
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, United States
| | - Melissa E. Petersen
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, United States
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, United states
| | - Sid E. O’Bryant
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, United states
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Robyn A. Honea
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, United States
| | - Jeffrey M. Burns
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, United States
| |
Collapse
|
29
|
Ennis GE, Saelzler U, Umpierrez GE, Moffat SD. Prediabetes and working memory in older adults. Brain Neurosci Adv 2020; 4:2398212820961725. [PMID: 33088921 PMCID: PMC7545783 DOI: 10.1177/2398212820961725] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/30/2020] [Indexed: 12/21/2022] Open
Abstract
Insulin sensitivity, pancreatic β-cell function, fasting glucose, and 2-h post-load glucose were related to cognition in cognitively healthy nondiabetic older adults. Thirty-five adults (⩾65 years) underwent a 2-h oral glucose tolerance test and cognitive testing. Seventeen had normal glucose tolerance and 18 had intermediate hyperglycaemia or prediabetes (World Health Organization criteria). Fasting glucose and 2-h post-load glucose and oral glucose tolerance test–derived measures of β-cell function (oral disposition index) and insulin sensitivity were analysed as predictors of four cognitive domains: verbal episodic memory, verbal fluency, executive function, and working memory. The prediabetes group had significantly worse working memory performance than the normal glucose tolerance group. Controlling for age and education, decreased oral disposition index, and increased 2-h post-load glucose were significantly related to worse working memory performance. Prediabetes may worsen working memory in healthy older adults. Reduced pancreatic β-cell function should be investigated as a contributor to age-related cognitive decline.
Collapse
Affiliation(s)
- Gilda E Ennis
- School of Psychology, College of Sciences, Georgia Institute of Technology, Atlanta, GA, USA.,Division of Geriatrics and Gerontology, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Ursula Saelzler
- School of Psychology, College of Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Guillermo E Umpierrez
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Scott D Moffat
- School of Psychology, College of Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
30
|
Mejido DC, Peny JA, Vieira MN, Ferreira ST, De Felice FG. Insulin and leptin as potential cognitive enhancers in metabolic disorders and Alzheimer's disease. Neuropharmacology 2020; 171:108115. [PMID: 32344008 DOI: 10.1016/j.neuropharm.2020.108115] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 03/30/2020] [Accepted: 04/20/2020] [Indexed: 02/08/2023]
|
31
|
Gonçalves RA, Wijesekara N, Fraser PE, De Felice FG. Behavioral Abnormalities in Knockout and Humanized Tau Mice. Front Endocrinol (Lausanne) 2020; 11:124. [PMID: 32226410 PMCID: PMC7080660 DOI: 10.3389/fendo.2020.00124] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 02/24/2020] [Indexed: 12/30/2022] Open
Abstract
Microtubule-associated protein tau assists in stabilizing microtubules and has been particularly implicated in Alzheimer's disease (AD). Given the importance of tau to AD pathogenesis and therapies, it is important to understand non-classic physiological functions for this protein inside and outside the central nervous system (CNS). Our group has previously shown that tau ablation triggers glucose intolerance and pancreatic dysfunction in mice, suggesting that tau plays a role in peripheral metabolic regulation. Little is known about the role of tau in anxiety. Moreover, inconsistent results have been generated regarding the effects of tau deletion in memory. Here, we characterize systemic insulin resistance, anxiety-related behavior and memory in 15 to 20 weeks old Wild-Type (WT), Tau knockout (TauKO) and a distinct hTau mouse model consisting of tau knockout expressing the longest isoform (2N4R) of a non-mutant WT human Tau protein under the prion promoter (hTau). Our findings demonstrate that tau deletion leads to anxiety-related behavior, impaired contextual and cued fear memory. The presence of a human Tau transgene did not ameliorate the phenotypes observed in animals lacking the mouse tau protein and it elicited impairments in learning, memory, and peripheral insulin sensitivity. Our results suggest that tau protein plays a role in memory and anxiety-related behavior. Our findings also indicate that previously unrecognized functions for tau protein may be a complicating factor in using animal models on the TauKO background. Understanding the link between tau pathophysiology and cognitive and metabolic alterations is of great importance to establish the complete contribution of tau protein to AD pathogenesis.
Collapse
Affiliation(s)
- Rafaella Araujo Gonçalves
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Nadeeja Wijesekara
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Paul E. Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- *Correspondence: Paul E. Fraser
| | - Fernanda G. De Felice
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
- Department of Psychiatry, Queen's University, Kingston, ON, Canada
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Fernanda G. De Felice
| |
Collapse
|
32
|
McNay EC, Pearson-Leary J. GluT4: A central player in hippocampal memory and brain insulin resistance. Exp Neurol 2020; 323:113076. [PMID: 31614121 PMCID: PMC6936336 DOI: 10.1016/j.expneurol.2019.113076] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/19/2019] [Accepted: 10/01/2019] [Indexed: 12/24/2022]
Abstract
Insulin is now well-established as playing multiple roles within the brain, and specifically as regulating hippocampal cognitive processes and metabolism. Impairments to insulin signaling, such as those seen in type 2 diabetes and Alzheimer's disease, are associated with brain hypometabolism and cognitive impairment, but the mechanisms of insulin's central effects are not determined. Several lines of research converge to suggest that the insulin-responsive glucose transporter GluT4 plays a central role in hippocampal memory processes, and that reduced activation of this transporter may underpin the cognitive impairments seen as a consequence of insulin resistance.
Collapse
Affiliation(s)
- Ewan C McNay
- Behavioral Neuroscience, University at Albany, Albany, NY, USA.
| | - Jiah Pearson-Leary
- Department of Anesthesiology, Abramson Research Center, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
33
|
Titova OE, Lindberg E, Tan X, Elmståhl S, Lind L, Schiöth HB, Benedict C. Association between sleep duration and executive function differs between diabetic and non-diabetic middle-aged and older adults. Psychoneuroendocrinology 2020; 111:104472. [PMID: 31610410 DOI: 10.1016/j.psyneuen.2019.104472] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 10/02/2019] [Accepted: 10/02/2019] [Indexed: 01/26/2023]
Abstract
Executive function is defined as a set of cognitive skills that are necessary to plan, monitor, and execute a sequence of goal-directed complex actions. Executive function is influenced by a variety of factors, including habitual sleep duration and diabetes. In the present study, we investigated in 18,769 Swedish adults (mean age: 61 y) the association between executive function, diabetes, and self-reported sleep duration. We observed a significant interaction between diabetes and sleep duration for the Trail Making Test (TMT) ratio (P < 0.01). This ratio is a measure of executive function where higher values indicate worse performance. Among diabetic participants (n = 1,523), long (defined as ≥9 h per day) vs. normal sleep duration (defined as 7-8 hours per day) was associated with a higher TMT ratio (P < 0.05). Similar significant results were observed in diabetic individuals without pharmacological treatment for diabetes (n = 1,062). Among non-diabetic participants (n = 17,246), no association between long sleep duration and the TMT ratio was observed (P > 0.05). Instead, short (defined as <7 h per day) vs. normal sleep duration was linked to a higher TMT ratio (P < 0.05). These findings suggest that the association between sleep duration and executive function differs between diabetic and non-diabetic middle-aged and older adults. Based on the cross-sectional design of the study, no firm conclusions can be drawn on the causality of the relations.
Collapse
Affiliation(s)
- Olga E Titova
- Department of Neuroscience, Uppsala University, Uppsala, Sweden; Department of Surgical Sciences, Section of Orthopedics, Uppsala University, Uppsala, Sweden.
| | - Eva Lindberg
- Department of Medical Sciences, Respiratory, Allergy and Sleep Research, Uppsala University, Uppsala, Sweden
| | - Xiao Tan
- Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Sölve Elmståhl
- Division of Geriatric Medicine, Department of Health Sciences, Clinical Research Centre (CRC), Lund University, Skåne University Hospital, Malmö, Sweden
| | - Lars Lind
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala, Sweden
| | - Helgi B Schiöth
- Department of Neuroscience, Uppsala University, Uppsala, Sweden; Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow, Russia
| | | |
Collapse
|
34
|
Yokomichi H, Kondo K, Nagamine Y, Yamagata Z, Kondo N. Dementia risk by combinations of metabolic diseases and body mass index: Japan Gerontological Evaluation Study Cohort Study. J Diabetes Investig 2020; 11:206-215. [PMID: 31207179 PMCID: PMC6944839 DOI: 10.1111/jdi.13103] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 05/31/2019] [Accepted: 06/13/2019] [Indexed: 01/01/2023] Open
Abstract
AIMS/INTRODUCTION To compare the dementia risk associated with pre-existing diabetes, hypertension, dyslipidemia, obesity (body mass index [BMI] ≥25 kg/m2 ) and underweight (BMI <18.5 kg/m2 ) among older adults. We also explored the dementia risk associated with combinations of metabolic diseases and BMI. MATERIALS AND METHODS We used data from the Japan Gerontological Evaluation Study. Participants completed a health checkup in 2010 and were followed for 5.8 years on average. Dementia was measured by municipal long-term care insurance registration. Diabetes, hypertension, dyslipidemia, obesity and underweight were diagnosed by medication use or health examination results. We calculated the incidence of dementia and adjusted hazard ratios (HRs). RESULTS Among 3,696 participating older adults, 338 developed dementia. Adjusted HRs (95% confidence intervals) in men and women (reference: those without corresponding disease of normal weight) were as follows: 2.22 (1.26-3.90) and 2.00 (1.07-3.74) for diabetes; 0.56 (0.29-1.10) and 1.05 (0.64-1.71) for hypertension; 1.30 (0.87-1.94) and 0.73 (0.49-1.08) for dyslipidemia; 0.73 (0.42-1.28) and 0.82 (0.49-1.37) for BMI of 25-29.9 kg/m2 ; and 1.04 (0.51-2.10) and 1.72 (1.05-2.81) for underweight. Dementia risk was significantly higher in underweight men with dyslipidemia (HR 4.15, 95% CI 1.79-9.63) compared with normal-weight men without dyslipidemia, and in underweight women with hypertension (HR 3.79, 1.55-9.28) compared with normal-weight women without hypertension. Dementia incidence was highest among underweight older adults with hypertension followed by dyslipidemia. CONCLUSIONS Among Japanese older adults, underweight and prevalent diabetes are risk factors for developing dementia. Lower BMI is also associated with a higher incidence of dementia.
Collapse
Affiliation(s)
| | - Katsunori Kondo
- Department of Social Preventive Medical SciencesCenter for Preventive Medical SciencesChiba UniversityChibaJapan
- Department of Gerontological EvaluationCenter for Gerontology and Social ScienceNational Center for Geriatrics and GerontologyAichiJapan
| | - Yuiko Nagamine
- Department of Social Preventive Medical SciencesCenter for Preventive Medical SciencesChiba UniversityChibaJapan
| | - Zentaro Yamagata
- Department of Health SciencesUniversity of YamanashiYamanashiJapan
| | - Naoki Kondo
- Department of Health Education and Health SociologySchool of Public HealthThe University of TokyoTokyoJapan
| |
Collapse
|
35
|
Smith PJ, Mabe SM, Sherwood A, Doraiswamy PM, Welsh-Bohmer KA, Burke JR, Kraus WE, Lin PH, Browndyke JN, Babyak MA, Hinderliter AL, Blumenthal JA. Metabolic and Neurocognitive Changes Following Lifestyle Modification: Examination of Biomarkers from the ENLIGHTEN Randomized Clinical Trial. J Alzheimers Dis 2020; 77:1793-1803. [PMID: 32925039 PMCID: PMC9999371 DOI: 10.3233/jad-200374] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Previous studies have demonstrated that aerobic exercise (AE) and the Dietary Approaches to Stop Hypertension (DASH) diet can improve neurocognition. However, the mechanisms by which lifestyle improves neurocognition have not been widely studied. We examined the associations between changes in metabolic, neurotrophic, and inflammatory biomarkers with executive functioning among participants from the Exercise and Nutritional Interventions for Neurocognitive Health Enhancement (ENLIGHTEN) trial. OBJECTIVE To examine the association between changes in metabolic function and neurocognition among older adults with cognitive impairment, but without dementia (CIND) participating in a comprehensive lifestyle intervention. METHODS ENLIGHTEN participants were randomized using a 2×2 factorial design to receive AE, DASH, both AE+DASH, or a health education control condition (HE) for six months. Metabolic biomarkers included insulin resistance (homeostatic model assessment [HOMA-IR]), leptin, and insulin-like growth factor (IGF-1); neurotrophic biomarkers included brain derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF); and inflammatory biomarkers included interleukin-6 (IL-6) and C-Reactive Protein (CRP). RESULTS Participants included 132 sedentary older adults (mean age = 65 [SD = 7]) with CIND. Results demonstrated that both AE (d = 0.48, p = 0.015) and DASH improved metabolic function (d = 0.37, p = 0.039), without comparable improvements in neurotrophic or inflammatory biomarkers. Greater improvements in metabolic function, including reduced HOMA-IR (B = -2.3 [-4.3, -0.2], p = 0.033) and increased IGF-1 (B = 3.4 [1.2, 5.7], p = 0.004), associated with increases in Executive Function. CONCLUSION Changes in neurocognition after lifestyle modification are associated with improved metabolic function.
Collapse
Affiliation(s)
- Patrick J Smith
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Stephanie M Mabe
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Andrew Sherwood
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - P Murali Doraiswamy
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Kathleen A Welsh-Bohmer
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - James R Burke
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - William E Kraus
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Pao-Hwa Lin
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Jeffrey N Browndyke
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Michael A Babyak
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Alan L Hinderliter
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - James A Blumenthal
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
36
|
Iozzo P, Guzzardi MA. Imaging of brain glucose uptake by PET in obesity and cognitive dysfunction: life-course perspective. Endocr Connect 2019; 8:R169-R183. [PMID: 31590145 PMCID: PMC6865363 DOI: 10.1530/ec-19-0348] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 10/07/2019] [Indexed: 12/17/2022]
Abstract
The prevalence of obesity has reached epidemic proportions and keeps growing. Obesity seems implicated in the pathogenesis of cognitive dysfunction, Alzheimer's disease and dementia, and vice versa. Growing scientific efforts are being devoted to the identification of central mechanisms underlying the frequent association between obesity and cognitive dysfunction. Glucose brain handling undergoes dynamic changes during the life-course, suggesting that its alterations might precede and contribute to degenerative changes or signaling abnormalities. Imaging of the glucose analog 18F-labeled fluorodeoxyglucose (18FDG) by positron emission tomography (PET) is the gold-standard for the assessment of cerebral glucose metabolism in vivo. This review summarizes the current literature addressing brain glucose uptake measured by PET imaging, and the effect of insulin on brain metabolism, trying to embrace a life-course vision in the identification of patterns that may explain (and contribute to) the frequent association between obesity and cognitive dysfunction. The current evidence supports that brain hypermetabolism and brain insulin resistance occur in selected high-risk conditions as a transient phenomenon, eventually evolving toward normal or low values during life or disease progression. Associative studies suggest that brain hypermetabolism predicts low BDNF levels, hepatic and whole body insulin resistance, food desire and an unfavorable balance between anticipated reward from food and cognitive inhibitory control. Emerging mechanistic links involve the microbiota and the metabolome, which correlate with brain metabolism and cognition, deserving attention as potential future prevention targets.
Collapse
Affiliation(s)
- Patricia Iozzo
- Institute of Clinical Physiology, National Research Council (CNR), Pisa, Italy
- Correspondence should be addressed to P Iozzo:
| | | |
Collapse
|
37
|
Ahmed A, Zeng G, Jiang D, Lin H, Azhar M, Farooq AD, Choudhary MI, Liu X, Wang Q. Time-dependent impairments in learning and memory in Streptozotocin-induced hyperglycemic rats. Metab Brain Dis 2019; 34:1431-1446. [PMID: 31286327 DOI: 10.1007/s11011-019-00448-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 06/10/2019] [Indexed: 12/17/2022]
Abstract
The sedentary lifestyle is responsible for the high prevalence of diabetes which also impairs cognition including learning and memory. Various studies have highlighted the learning and memory impairments in rodent models but data regarding the timeline of their development and their correlation to biochemical parameters are scarce. So, the present study was designed to investigate the type of memory which is more susceptible to hyperglycemia and its correlation with biochemical parameters such as inflammatory cytokines, cAMP response element binding (CREB) and protein kinase B (Akt) activation. Hyperglycemia was induced using streptozotocin (STZ, 45 mg/kg i.p.) and confirmed by measuring fasting blood glucose levels after 1 week of STZ injection. Learning and memory deficits were evaluated using the Novel Object Recognition Test (NORT) and Morris water maze (MWM), and correlated with biochemical parameters (TNF-α, IL-1β, and dopamine) at 3, 6 and 9 weeks. STZ-injected rats after 3 weeks of injection demonstrated moderate hyperglycemia (blood glucose = 7.99 ± 0.62 mM) with intact learning and reference memory; however, their working memory was impaired in MWM. Severe hyperglycemia (blood glucose = 11.51 ± 0.69 mM) accompanied by impaired short, long, and working memory was evident after 6 weeks whereas learning was intact. After 9 weeks of STZ injection, hyperglycemia was more pronounced (13.69 ± 1.43 mM) and accompanied by a learning deficit in addition to short, long, and working memory impairments. The extent of hyperglycemia either in terms of duration or severity resulted in enhanced inflammation, down-regulation of the level of dopamine, protein expression of AKT and CREB, which possibly affected learning and memory negatively.
Collapse
Affiliation(s)
- Ayaz Ahmed
- Affiliated TCM hospital/ Sino-Portugal TCM International Cooperation Center / Department of Physiology in School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
- Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, China
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Guirong Zeng
- Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, China
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dejiang Jiang
- Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, China
| | - Haiying Lin
- Affiliated TCM hospital/ Sino-Portugal TCM International Cooperation Center / Department of Physiology in School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Mudassar Azhar
- Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, China
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Ahsana Dar Farooq
- Hamdard Al-Majeed College of Eastern Medicine, Hamdard University, Karachi, 74600, Pakistan
| | - Muhammad Iqbal Choudhary
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
- Department of Biochemistry, College of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Xinmin Liu
- Affiliated TCM hospital/ Sino-Portugal TCM International Cooperation Center / Department of Physiology in School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China.
- Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, China.
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Qiong Wang
- Affiliated TCM hospital/ Sino-Portugal TCM International Cooperation Center / Department of Physiology in School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
38
|
Beyer F, Kharabian Masouleh S, Kratzsch J, Schroeter ML, Röhr S, Riedel-Heller SG, Villringer A, Witte AV. A Metabolic Obesity Profile Is Associated With Decreased Gray Matter Volume in Cognitively Healthy Older Adults. Front Aging Neurosci 2019; 11:202. [PMID: 31427957 PMCID: PMC6688742 DOI: 10.3389/fnagi.2019.00202] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 07/17/2019] [Indexed: 12/22/2022] Open
Abstract
Obesity is a risk factor for cognitive decline and gray matter volume loss in aging. Studies have shown that different metabolic factors, e.g., dysregulated glucose metabolism and systemic inflammation, might mediate this association. Yet, even though these risk factors tend to co-occur, they have mostly been investigated separately, making it difficult to establish their joint contribution to gray matter volume structure in aging. Here, we therefore aimed to determine a metabolic profile of obesity that takes into account different anthropometric and metabolic measures to explain differences in gray matter volume in aging. We included 748 elderly, cognitively healthy participants (age range: 60 - 79 years, BMI range: 17 - 42 kg/m2) of the LIFE-Adult Study. All participants had complete information on body mass index, waist-to-hip ratio, glycated hemoglobin, total blood cholesterol, high-density lipoprotein, interleukin-6, C-reactive protein, adiponectin and leptin. Voxelwise gray matter volume was extracted from T1-weighted images acquired on a 3T Siemens MRI scanner. We used partial least squares correlation to extract latent variables with maximal covariance between anthropometric, metabolic and gray matter volume and applied permutation/bootstrapping and cross-validation to test significance and reliability of the result. We further explored the association of the latent variables with cognitive performance. Permutation tests and cross-validation indicated that the first pair of latent variables was significant and reliable. The metabolic profile was driven by negative contributions from body mass index, waist-to-hip ratio, glycated hemoglobin, C-reactive protein and leptin and a positive contribution from adiponectin. It positively covaried with gray matter volume in temporal, frontal and occipital lobe as well as subcortical regions and cerebellum. This result shows that a metabolic profile characterized by high body fat, visceral adiposity and systemic inflammation is associated with reduced gray matter volume and potentially reduced executive function in older adults. We observed the highest contributions for body weight and fat mass, which indicates that factors underlying sustained energy imbalance, like sedentary lifestyle or intake of energy-dense food, might be important determinants of gray matter structure in aging.
Collapse
Affiliation(s)
- Frauke Beyer
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Subproject A1, CRC 1052 “Obesity Mechanisms”, University of Leipzig, Leipzig, Germany
| | - Shahrzad Kharabian Masouleh
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Institute of Neuroscience and Medicine (INM-7: Brain and Behaviour), Research Centre Jülich, Jülich, Germany
| | - Jürgen Kratzsch
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig, Leipzig, Germany
| | - Matthias L. Schroeter
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Clinic for Cognitive Neurology, University of Leipzig, Leipzig, Germany
| | - Susanne Röhr
- Institute of Social Medicine, Occupational Health and Public Health (ISAP), University of Leipzig, Leipzig, Germany
| | - Steffi G. Riedel-Heller
- Institute of Social Medicine, Occupational Health and Public Health (ISAP), University of Leipzig, Leipzig, Germany
| | - Arno Villringer
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Subproject A1, CRC 1052 “Obesity Mechanisms”, University of Leipzig, Leipzig, Germany
- Clinic for Cognitive Neurology, University of Leipzig, Leipzig, Germany
| | - A. Veronica Witte
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Subproject A1, CRC 1052 “Obesity Mechanisms”, University of Leipzig, Leipzig, Germany
- Clinic for Cognitive Neurology, University of Leipzig, Leipzig, Germany
| |
Collapse
|
39
|
Hugenschmidt CE, Leng X, Lyles M, Michael L, Dougherty A, Babcock P, Baker LD, Brinkley TE, Nicklas BJ. Cognitive Effects of Adding Caloric Restriction to Aerobic Exercise Training in Older Adults with Obesity. Obesity (Silver Spring) 2019; 27:1266-1274. [PMID: 31199592 PMCID: PMC6656607 DOI: 10.1002/oby.22525] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 04/16/2019] [Indexed: 12/17/2022]
Abstract
OBJECTIVE This study examined the short- and long-term effects of adding caloric restriction to 5 months of aerobic exercise training on executive function in sedentary older adults with obesity. METHODS Sedentary adults with obesity aged 65 to 79 years completed a randomized trial investigating the cardiorespiratory benefits of adding moderate (~ 250 kcal) or high (~ 600 kcal) caloric restriction to a 20-week aerobic exercise program. Approximately half (n = 88) completed a cognitive assessment battery at baseline, post intervention, and 18 to 24 months after intervention completion. The primary outcome was an executive function composite score. RESULTS In the overall sample, the executive function composite increased 0.114 from baseline to postintervention (P = 0.01). Randomization to caloric restriction did not significantly alter executive function over aerobic exercise alone, nor were there between-group differences on any individual executive function test following the intervention or at long-term follow-up. Adding caloric restriction to exercise was associated with a modest increase in Mini-Mental State Examination score (P = 0.04). In the overall sample, increases from baseline at long-term follow-up were noted in digit symbol and word list recall performance as well. CONCLUSIONS Adding caloric restriction to a 20-week aerobic exercise program does not worsen or improve executive function more than exercise alone assessed up to 24 months post randomization.
Collapse
Affiliation(s)
- Christina E. Hugenschmidt
- Wake Forest School of Medicine, Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Winston-Salem, NC 27157, USA
| | - Xiaoyan Leng
- Wake Forest School of Medicine, Department of Biostatistical Sciences, Winston-Salem, NC 27157, USA
| | - Mary Lyles
- Wake Forest School of Medicine, Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Winston-Salem, NC 27157, USA
| | - Lemaat Michael
- Wake Forest School of Medicine, Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Winston-Salem, NC 27157, USA
| | - Ashley Dougherty
- Duke University Medical Center, Family Medicine, Durham, NC 27705, USA
| | - Phyllis Babcock
- Wake Forest School of Medicine, Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Winston-Salem, NC 27157, USA
| | - Laura D. Baker
- Wake Forest School of Medicine, Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Winston-Salem, NC 27157, USA
| | - Tina E. Brinkley
- Wake Forest School of Medicine, Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Winston-Salem, NC 27157, USA
| | - Barbara J. Nicklas
- Wake Forest School of Medicine, Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
40
|
Lee TH, Hurwitz EL, Cooney RV, Wu YY, Wang CY, Masaki K, Grandinetti A. Late life insulin resistance and Alzheimer's disease and dementia: The Kuakini Honolulu heart program. J Neurol Sci 2019; 403:133-138. [DOI: 10.1016/j.jns.2019.06.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 06/03/2019] [Accepted: 06/27/2019] [Indexed: 01/24/2023]
|
41
|
Drummen M, Heinecke A, Dorenbos E, Vreugdenhil A, Raben A, Westerterp-Plantenga MS, Adam TC. Reductions in body weight and insulin resistance are not associated with changes in grey matter volume or cortical thickness during the PREVIEW study. J Neurol Sci 2019; 403:106-111. [PMID: 31255969 DOI: 10.1016/j.jns.2019.06.017] [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] [Received: 11/02/2018] [Revised: 05/10/2019] [Accepted: 06/13/2019] [Indexed: 02/05/2023]
Abstract
INTRODUCTION The effect of changes in body weight or insulin resistance on grey matter volume and cortical thickness change are unclear. The present observational study assessed effects of an 8-week weight loss period (≥8% of body weight), and a subsequent 22-month weight maintenance period on grey matter volume and cortical thickness. METHODS A total of 24 participants (12f/12 m; age 52.8 ± 10.6 years) with overweight/obesity and pre-diabetes were recruited. T1-weighted magnetic resonance imaging was used to determine grey matter volume and cortical thickness at baseline, after the weight loss period and after a medium to high dietary protein weight maintenance period. RESULTS At baseline, global grey matter volume was inversely associated with HOMA-IR, adjusted for sex and age (r = -0.42; p = .049). During the weight loss period participants decreased their BMI (32.1 ± 3.3 to 28.1 ± 2.8 kg/m2, p < .01), body-fat (41.6 ± 6.4 to 35.0 ± 8.0%, p < .01) and insulin resistance (HOMA-IR: 4.0 ± 2.0 to 1.8 ± 0.9, p < .01). During the 22-month weight maintenance period, these parameters gradually increased again (BMI: 29.3 ± 3.8 kg/m2; body-fat: 37.8 ± 9.3%; HOMA-IR: 2.9 ± 1.4, p < .01). Global grey matter volume and cortical thickness did not change significantly during the weight loss or weight maintenance period. Changes in body weight, body-fat percentage or insulin sensitivity were not associated with changes in global grey matter volume. CONCLUSION In conclusion, we confirmed that global grey brain matter volume was inversely associated with insulin resistance at baseline, yet an intervention yielding a decrease in insulin resistance did not lead to changes in global grey brain matter volume or cortical thickness. TRIAL REGISTRATION The trial is registered with ClinicalTrials.gov, NCT01777893.
Collapse
Affiliation(s)
- M Drummen
- Department of Nutrition and Movement Sciences, Maastricht University Medical Centre +, Maastricht, the Netherlands; NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht, Maastricht University, the Netherlands.
| | - A Heinecke
- Department of Cognitive Neuroscience, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - E Dorenbos
- Centre for Overweight Adolescent and Children's Health Care (COACH), Department of Paediatrics, Maastricht University Medical Centre +, the Netherlands; NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht, Maastricht University, the Netherlands
| | - A Vreugdenhil
- Centre for Overweight Adolescent and Children's Health Care (COACH), Department of Paediatrics, Maastricht University Medical Centre +, the Netherlands; NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht, Maastricht University, the Netherlands
| | - A Raben
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - M S Westerterp-Plantenga
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht, Maastricht University, the Netherlands
| | - T C Adam
- Department of Nutrition and Movement Sciences, Maastricht University Medical Centre +, Maastricht, the Netherlands; NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht, Maastricht University, the Netherlands
| |
Collapse
|
42
|
Frangou S, Shirali M, Adams MJ, Howard DM, Gibson J, Hall LS, Smith BH, Padmanabhan S, Murray AD, Porteous DJ, Haley CS, Deary IJ, Clarke TK, McIntosh AM. Insulin resistance: Genetic associations with depression and cognition in population based cohorts. Exp Neurol 2019; 316:20-26. [PMID: 30965038 PMCID: PMC6503941 DOI: 10.1016/j.expneurol.2019.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/26/2019] [Accepted: 04/03/2019] [Indexed: 01/07/2023]
Abstract
Insulin resistance, broadly defined as the reduced ability of insulin to exert its biological action, has been associated with depression and cognitive dysfunction in observational studies. However, it is unclear whether these associations are causal and whether they might be underpinned by other shared factors. To address this knowledge gap, we capitalized on the stability of genetic biomarkers through the lifetime, and on their unidirectional relationship with depression and cognition. Specifically, we determined the association between quantitative measures of cognitive function and depression and genetic instruments of insulin resistance traits in two large-scale population samples, the Generation Scotland: Scottish Family Health Study (GS: SFHS; N = 19,994) and in the UK Biobank (N = 331,374). In the GS:SFHS, the polygenic risk score (PRS) for fasting insulin was associated with verbal intelligence and depression while the PRS for the homeostasis model assessment of insulin resistance was associated with verbal intelligence. Despite this overlap in genetic architecture, Mendelian randomization analyses in the GS:SFHS and in the UK Biobank samples did not yield evidence for causal associations from insulin resistance traits to either depression or cognition. These findings may be due to weak genetic instruments, limited cognitive measures and insufficient power but they may also indicate the need to identify other biological mechanisms that may mediate the relationship from insulin resistance to depression and cognition.
Collapse
Affiliation(s)
- Sophia Frangou
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Masoud Shirali
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Mark J Adams
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - David M Howard
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Jude Gibson
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Lynsey S Hall
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Blair H Smith
- Division of Population Health Sciences, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Sandosh Padmanabhan
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Alison D Murray
- Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen, UK
| | - David J Porteous
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK; Generation Scotland, Medical Genetics Section, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Chris S Haley
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK; Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Toni-Kim Clarke
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Andrew M McIntosh
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK; Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK; Department of Psychology, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
43
|
Arnoriaga Rodríguez M, Blasco G, Coll C, Biarnés C, Contreras-Rodríguez O, Garre-Olmo J, Puig J, Gich J, Ricart W, Ramió-Torrentà L, Fernández-Real JM. Glycated Hemoglobin, but not Insulin Sensitivity, is Associated with Memory in Subjects with Obesity. Obesity (Silver Spring) 2019; 27:932-942. [PMID: 30985999 DOI: 10.1002/oby.22457] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/24/2019] [Indexed: 01/21/2023]
Abstract
OBJECTIVE Obesity has been related to later-life dementia. Serum glucose levels and insulin resistance are known to influence cognition in individuals with diabetes. This study aimed to evaluate memory function in middle-aged individuals with obesity in association with glucose metabolism and brain iron content. METHODS This was a cross-sectional case-control study including 121 participants aged 27.2 to 66.6 years (56 without obesity, 65 with obesity) stratified according to sex and menopausal status. Insulin sensitivity, body composition, brain iron content, and memory function were evaluated by euglycemic hyperinsulinemic clamp, dual-energy x-ray absorptiometry, magnetic resonance relaxometry (R2*), and California Verbal Learning Test, respectively. RESULTS Women with obesity, but not men, had lower scores in some California Verbal Learning Tests in association with metabolic parameters and increased brain iron content compared with controls. Fasting plasma glucose, glycated hemoglobin (HbA1c; within normal range), and R2* were negatively associated with memory scores, whereas insulin sensitivity showed positive associations. Remarkably, only HbA1c levels and R2* in the right inferior fronto-orbital region remained significant after controlling for age, sex, education, and BMI. CONCLUSIONS Impairments in memory function in middle-aged women with obesity are associated with HbA1c levels and brain iron content independently of insulin sensitivity. These results may have implications in the design of therapeutic strategies in women with obesity.
Collapse
Affiliation(s)
- María Arnoriaga Rodríguez
- Department of Diabetes, Endocrinology and Nutrition, Girona Biomedical Research Institute, Dr. Josep Trueta University Hospital, Center for Physiopathology of Obesity and Nutrition, Girona, Spain
- Faculty of Medicine, Department of Medical Sciences, University of Girona, Girona, Spain
| | - Gerard Blasco
- Institute of Diagnostic Imaging-Research Unit, Parc Sanitari Pere Virgili, Barcelona, Spain
- Department of Medical Imaging, Girona Biomedical Research Institute, Girona, Spain
| | - Clàudia Coll
- Neuroimmunology and Multiple Sclerosis Unit, Department of Neurology, Dr. Josep Trueta University Hospital, Girona, Spain
| | - Carles Biarnés
- Department of Medical Imaging, Girona Biomedical Research Institute, Girona, Spain
| | - Oren Contreras-Rodríguez
- Department of Psychiatry, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL) and Biomedical Research Networking Center for Mental Health (CIBERSAM), L'Hospitalet de Llobregat, Spain
| | - Josep Garre-Olmo
- Faculty of Medicine, Department of Medical Sciences, University of Girona, Girona, Spain
- Research Group on Aging, Health and Disability, Girona Biomedical Research Institute, Health Assistance Institute, Girona, Spain
| | - Josep Puig
- Faculty of Medicine, Department of Medical Sciences, University of Girona, Girona, Spain
- Institute of Diagnostic Imaging-Research Unit, Parc Sanitari Pere Virgili, Barcelona, Spain
- Department of Medical Imaging, Girona Biomedical Research Institute, Girona, Spain
| | - Jordi Gich
- Faculty of Medicine, Department of Medical Sciences, University of Girona, Girona, Spain
- Neurodegeneration and Neuroinflammation Group, Girona Biomedical Research Institute, Girona, Spain
| | - Wifredo Ricart
- Department of Diabetes, Endocrinology and Nutrition, Girona Biomedical Research Institute, Dr. Josep Trueta University Hospital, Center for Physiopathology of Obesity and Nutrition, Girona, Spain
- Faculty of Medicine, Department of Medical Sciences, University of Girona, Girona, Spain
| | - Lluís Ramió-Torrentà
- Faculty of Medicine, Department of Medical Sciences, University of Girona, Girona, Spain
- Neuroimmunology and Multiple Sclerosis Unit, Department of Neurology, Dr. Josep Trueta University Hospital, Girona, Spain
- Neurodegeneration and Neuroinflammation Group, Girona Biomedical Research Institute, Girona, Spain
| | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Girona Biomedical Research Institute, Dr. Josep Trueta University Hospital, Center for Physiopathology of Obesity and Nutrition, Girona, Spain
- Faculty of Medicine, Department of Medical Sciences, University of Girona, Girona, Spain
| |
Collapse
|
44
|
Hari Dass SA, McCracken K, Pokhvisneva I, Chen LM, Garg E, Nguyen TTT, Wang Z, Barth B, Yaqubi M, McEwen LM, MacIsaac JL, Diorio J, Kobor MS, O'Donnell KJ, Meaney MJ, Silveira PP. A biologically-informed polygenic score identifies endophenotypes and clinical conditions associated with the insulin receptor function on specific brain regions. EBioMedicine 2019; 42:188-202. [PMID: 30922963 PMCID: PMC6491717 DOI: 10.1016/j.ebiom.2019.03.051] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Activation of brain insulin receptors modulates reward sensitivity, inhibitory control and memory. Variations in the functioning of this mechanism likely associate with individual differences in the risk for related mental disorders (attention deficit hyperactivity disorder or ADHD, addiction, dementia), in agreement with the high co-morbidity between insulin resistance and psychopathology. These neurobiological mechanisms can be explored using genetic studies. We propose a novel, biologically informed genetic score reflecting the mesocorticolimbic and hippocampal insulin receptor-related gene networks, and investigate if it predicts endophenotypes (impulsivity, cognitive ability) in community samples of children, and psychopathology (addiction, dementia) in adults. METHODS Lists of genes co-expressed with the insulin receptor in the mesocorticolimbic system or hippocampus were created. SNPs from these genes (post-clumping) were compiled in a polygenic score using the association betas described in a conventional GWAS (ADHD in the mesocorticolimbic score and Alzheimer in the hippocampal score). Across multiple samples (n = 4502), the biologically informed, mesocorticolimbic or hippocampal specific insulin receptor polygenic scores were calculated, and their ability to predict impulsivity, risk for addiction, cognitive performance and presence of Alzheimer's disease was investigated. FINDINGS The biologically-informed ePRS-IR score showed better prediction of child impulsivity and cognitive performance, as well as risk for addiction and Alzheimer's disease in comparison to conventional polygenic scores for ADHD, addiction and dementia. INTERPRETATION This novel, biologically-informed approach enables the use of genomic datasets to probe relevant biological processes involved in neural function and disorders. FUND: Toxic Stress Research network of the JPB Foundation, Jacobs Foundation (Switzerland), Sackler Foundation.
Collapse
Affiliation(s)
- Shantala A Hari Dass
- Sackler Institute for Epigenetics & Psychobiology, McGill University, 6875 Boulevard LaSalle, Verdun, QC H4H 1R3, Canada
| | - Kathryn McCracken
- John Abbott College, Sainte-Anne-de-Bellevue, 6875 Boulevard LaSalle, Verdun, QC H4H 1R3, Canada; McGill University, 6875 Boulevard LaSalle, Verdun, QC H4H 1R3, Canada
| | - Irina Pokhvisneva
- Sackler Institute for Epigenetics & Psychobiology, McGill University, 6875 Boulevard LaSalle, Verdun, QC H4H 1R3, Canada
| | - Lawrence M Chen
- Sackler Institute for Epigenetics & Psychobiology, McGill University, 6875 Boulevard LaSalle, Verdun, QC H4H 1R3, Canada
| | - Elika Garg
- Sackler Institute for Epigenetics & Psychobiology, McGill University, 6875 Boulevard LaSalle, Verdun, QC H4H 1R3, Canada
| | - Thao T T Nguyen
- Sackler Institute for Epigenetics & Psychobiology, McGill University, 6875 Boulevard LaSalle, Verdun, QC H4H 1R3, Canada
| | - Zihan Wang
- Sackler Institute for Epigenetics & Psychobiology, McGill University, 6875 Boulevard LaSalle, Verdun, QC H4H 1R3, Canada
| | - Barbara Barth
- McGill University, 6875 Boulevard LaSalle, Verdun, QC H4H 1R3, Canada
| | - Moein Yaqubi
- McGill University, 6875 Boulevard LaSalle, Verdun, QC H4H 1R3, Canada
| | - Lisa M McEwen
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, Department of Medical Genetics, The University of British Columbia, 938 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada
| | - Julie L MacIsaac
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, Department of Medical Genetics, The University of British Columbia, 938 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada
| | - Josie Diorio
- Sackler Institute for Epigenetics & Psychobiology, McGill University, 6875 Boulevard LaSalle, Verdun, QC H4H 1R3, Canada
| | - Michael S Kobor
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, Department of Medical Genetics, The University of British Columbia, 938 West 28th Avenue, Vancouver, BC V5Z 4H4, Canada
| | - Kieran J O'Donnell
- Department of Psychiatry, Faculty of Medicine, McGill University, 6875 Boulevard LaSalle, Verdun, QC H4H 1R3, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, 6875 Boulevard LaSalle, Verdun, QC H4H 1R3, Canada; Sackler Institute for Epigenetics & Psychobiology, McGill University, 6875 Boulevard LaSalle, Verdun, QC H4H 1R3, Canada
| | - Michael J Meaney
- Department of Psychiatry, Faculty of Medicine, McGill University, 6875 Boulevard LaSalle, Verdun, QC H4H 1R3, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, 6875 Boulevard LaSalle, Verdun, QC H4H 1R3, Canada; Sackler Institute for Epigenetics & Psychobiology, McGill University, 6875 Boulevard LaSalle, Verdun, QC H4H 1R3, Canada; Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Brenner Centre for Molecular Medicine, 30 Medical Drive, 117609, Singapore
| | - Patricia P Silveira
- Department of Psychiatry, Faculty of Medicine, McGill University, 6875 Boulevard LaSalle, Verdun, QC H4H 1R3, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, 6875 Boulevard LaSalle, Verdun, QC H4H 1R3, Canada; Sackler Institute for Epigenetics & Psychobiology, McGill University, 6875 Boulevard LaSalle, Verdun, QC H4H 1R3, Canada.
| |
Collapse
|
45
|
Smith PJ, Mabe S, Sherwood A, Babyak MA, Murali Doraiswamy P, Welsh-Bohmer KA, Kraus W, Burke J, Hinderliter A, Blumenthal JA. Association Between Insulin Resistance, Plasma Leptin, and Neurocognition in Vascular Cognitive Impairment. J Alzheimers Dis 2019; 71:921-929. [PMID: 31476159 PMCID: PMC10840083 DOI: 10.3233/jad-190569] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Greater body weight has been associated impairments in neurocognition and greater dementia risk, although the mechanisms linking weight and neurocognition have yet to be adequately delineated. OBJECTIVE To examine metabolic mechanisms underlying the association between obesity and neurocognition. METHODS We conducted a secondary analysis of weight, neurocognition, and the potentially mediating role of metabolic and inflammatory biomarkers among 160 participants from the ENLIGHTEN trial of vascular cognitive impairment, no dementia (CIND). Neurocognition was assessed using a 45-minute assessment battery assessing Executive Function, Verbal and Visual Memory. We considered three metabolic biomarkers: insulin resistance (homeostatic model assessment [HOMA-IR]), plasma leptin, and insulin-like growth factor (IGF-1). Inflammation was assessed using C-reactive protein. Multiple regression analyses were used. RESULTS Participants included 160 sedentary older adults with CIND. Participants tended to be overweight or obese (mean BMI = 32.5 [SD = 4.8]). Women exhibited higher BMI (p = 0.043), CRP (p < 0.001), and leptin (p < 0.001) compared with men. Higher BMI levels were associated with worse performance on measures of Executive Function (β= -0.16, p = 0.024) and Verbal Memory (β= -0.16, p = 0.030), but not Visual Memory (β= 0.05, p = 0.500). Worse metabolic biomarker profiles also were associated with lower Executive Function (β= -0.12, p = 0.050). Mediation analyses suggested leptin was a plausible candidate as a mediator between BMI and Executive Function. CONCLUSIONS In overweight and obese adults with vascular CIND, the association between greater weight and poorer executive function may be mediated by higher leptin resistance.
Collapse
Affiliation(s)
- Patrick J. Smith
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Stephanie Mabe
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Andrew Sherwood
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Michael A. Babyak
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - P. Murali Doraiswamy
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Kathleen A. Welsh-Bohmer
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - William Kraus
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - James Burke
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Alan Hinderliter
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - James A. Blumenthal
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
46
|
Diffusion spectrum imaging in white matter microstructure in subjects with type 2 diabetes. PLoS One 2018; 13:e0203271. [PMID: 30427838 PMCID: PMC6235258 DOI: 10.1371/journal.pone.0203271] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 08/18/2018] [Indexed: 11/19/2022] Open
Abstract
This study aimed to investigate the feasibility and clinical applicability of Diffusion Spectrum Imaging (DSI) for quantitative detection of white matter microstructural integrity. Twenty-seven patients with type 2 diabetes mellitus (T2DM; aged 60.6±7.6 years) and 21 healthy controls (HC; aged 56.1±7.8 years) underwent high-resolution T1-weighted imaging and DSI scanning. Cognitive function scores were obtained using such instruments as the Montreal Cognitive Assessment (MoCA). The fasting blood glucose, glycated hemoglobin, and total cholesterol (CHO) of T2DM were measured. The bilateral uncinate fasciculus and superior cingulum bundle were reconstructed by DSI tractography. Statistical analysis was performed using SPSS 21.0 software and P<0.05 was considered significant. Generalized fractional anisotropy (GFA) values were significantly decreased in the left uncinate fasciculus (t = −2.915, p = 0.005) and right superior cingulum bundle (t = −2.604, p = 0.012) in T2DM patients compared with the healthy controls (p<0.05). The MoCA (t = −3.339, p = 0.002) and CDT (t = −3.039, p = 0.004) scores of T2DM were significantly lower than those of healthy controls. Meanwhile, the GFA value of the right superior cingulum bundle was negatively associated with VFT score (r = −0.475, p = 0.012), and that of the right superior cingulum bundle was negatively associated with blood CHO level (r = −0.458, p = 0.016). DSI tractography is capable of evaluating the microstructural integrity of the white matter bundle in T2DM and is related to clinical cognitive scores and related biochemical indices; therefore, it can help to predict early white matter abnormalities in T2DM.
Collapse
|
47
|
Lee J, Kim J, Shin SA, Park S, Yoon DH, Kim H, Kim YK, Moon MK, Koo BK, Lee JY. Moderating Effect of Insulin Resistance on the Relationship between Gray Matter Volumes and Cognitive Function. J Clin Med 2018; 7:jcm7110413. [PMID: 30400348 PMCID: PMC6262494 DOI: 10.3390/jcm7110413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 10/24/2018] [Accepted: 11/01/2018] [Indexed: 11/16/2022] Open
Abstract
Background: It is controversial whether exposure to insulin resistance accelerates cognitive deterioration. The present study aimed to investigate the association between insulin resistance and gray matter volume loss to predict the cognitive decline. Methods: We recruited 160 participants (78 with Alzheimer’s disease and 82 without Alzheimer’s disease). Insulin resistance, regional gray matter volume, and cognitive function were assessed. A hierarchical moderated multiple regression (MMR) model was used to determine any associations among insulin resistance, structural changes in the brain, and cognitive decline. Results: The volumes of 7 regions in the gray matter were negatively related to insulin resistance in Alzheimer’s disease (p =0.032). Hierarchical MMR analysis indicated that insulin resistance did not directly affect the cognitive decline but moderated the cognitive decline through the decrease in gray matter volume in the key brain regions, i.e., inferior orbitofrontal gyrus (left), middle cingulate gyrus (right), hippocampus (right), and precuneus (right) (p < 0.05 in each case). Conclusion: Insulin resistance appears to exacerbate the cognitive decline associated with several gray matter volume loss.
Collapse
Affiliation(s)
- Jiyeon Lee
- Department of Psychiatry, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul 07061, Korea.
| | - Jihyeon Kim
- College of Medicine, Seoul National University, Seoul 03080, Korea.
| | - Seong A Shin
- Department of Biomedical Sciences, Seoul National University, Seoul 08826, Korea.
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul 07061, Korea.
| | - Soowon Park
- Department of Education, Sejong University, Seoul 05006, Korea.
| | - Dong Hyun Yoon
- Department of Psychiatry and Behavioral Science, SMG-SNU Boramae Medical Center, Seoul 07061, Korea.
| | - Hongrae Kim
- Department of Psychiatry, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul 07061, Korea.
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul 07061, Korea.
| | - Min Kyong Moon
- Department of Internal Medicine, Seoul National University College of Medicine & SMG-SNU Boramae Medical Center, Seoul 07061, Korea.
| | - Bo Kyung Koo
- Department of Internal Medicine, Seoul National University College of Medicine & SMG-SNU Boramae Medical Center, Seoul 07061, Korea.
| | - Jun-Young Lee
- Department of Psychiatry and Neuroscience Research Institute, Seoul National University College of Medicine & SMG-SNU Boramae Medical Center, Seoul 07061, Korea.
| |
Collapse
|
48
|
Jones N, Riby LM, Smith MA. Glucose regulation and face recognition deficits in older adults: the role of attention. NEUROPSYCHOLOGY, DEVELOPMENT, AND COGNITION. SECTION B, AGING, NEUROPSYCHOLOGY AND COGNITION 2018; 25:673-694. [PMID: 28805109 DOI: 10.1080/13825585.2017.1365813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The present study investigated the perceptual, attentional, and memory processes underlying face recognition deficits observed in older adults with impaired glucoregulation. Participants were categorized as good glucoregulators or poor glucoregulators on the basis of an oral glucose tolerance test. Using event-related potential (ERP) methodology, 23 participants (62-88 years) performed a 2-stimulus oddball task. Participants were asked to rate and memorize 10 "target" faces, which were then presented amongst 120 unfamiliar foils. Behavioral results indicated that good glucoregulators were significantly more accurate at recognizing target faces. ERP markers of early visual perception (P1 and N170 components) and memory formation (P3 component) were unaffected by glucoregulatory efficiency. The P2 component, an index of attentional processing, was larger and delayed in the poor glucoregulators. To the best of our knowledge, this study is the first to suggest that face recognition deficits in poor glucoregulators may be due to impairments in attentional processing.
Collapse
Affiliation(s)
- Nicola Jones
- a Department of Psychology , Northumbria University , Newcastle upon Tyne , UK
- b Department of Psychology, Faculty of Science , Liverpool Hope University , Liverpool , UK
| | - Leigh M Riby
- a Department of Psychology , Northumbria University , Newcastle upon Tyne , UK
| | - Michael A Smith
- a Department of Psychology , Northumbria University , Newcastle upon Tyne , UK
- c Faculty of Health and Medical Sciences , University of Western Australia , Perth , Australia
| |
Collapse
|
49
|
Astrup A, Bügel S. Overfed but undernourished: recognizing nutritional inadequacies/deficiencies in patients with overweight or obesity. Int J Obes (Lond) 2018; 43:219-232. [PMID: 29980762 DOI: 10.1038/s41366-018-0143-9] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 04/17/2018] [Accepted: 05/20/2018] [Indexed: 02/06/2023]
Abstract
Overweight and obesity are highly prevalent throughout the world and can adversely affect the nutritional status of individuals. Studies have shown that many people with obesity have inadequate intake of iron, calcium, magnesium, zinc, copper, folate and vitamins A and B12, likely as a result of poor diet quality. Nutritional inadequacies or deficiencies may also occur due to altered pharmacokinetics in the individual with obesity and due to interactions in those with overweight or obesity with various pharmaceuticals. However, it has been demonstrated that the adult population in the United States as a whole is deficient in certain micronutrients as a result of the availability and overconsumption of high-calorie, low-nutrient processed foods. Poor nutrition may contribute to the development of certain chronic conditions, such as type 2 diabetes, which is already more prevalent in those with obesity. Clinicians need to be aware of these gaps, particularly in those individuals with obesity who are undergoing bariatric surgery or taking pharmaceutical products long term to facilitate weight loss. Patients with overweight or obesity likely struggle to achieve a balanced diet and may benefit from consultation with a dietitian. Along with providing recommendations for healthy eating and exercise, supplementation with specific micronutrients or multivitamins should be considered for individuals at the highest risk for or with established deficiencies. Further research is needed to understand the factors underlying nutritional inadequacies in individuals with overweight or obesity, as well as the outcomes of treatment strategies employed to address them.
Collapse
Affiliation(s)
- Arne Astrup
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark.
| | - Susanne Bügel
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
50
|
Sherzai AZ, Shaheen M, Yu JJ, Talbot K, Sherzai D. Insulin resistance and cognitive test performance in elderly adults: National health and nutrition examination survey (NHANES). J Neurol Sci 2018; 388:97-102. [DOI: 10.1016/j.jns.2017.11.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/15/2017] [Accepted: 11/22/2017] [Indexed: 01/18/2023]
|