1
|
Ye XW, Zhang HX, Li Q, Li CS, Zhao CJ, Xia LJ, Ren HM, Wang XX, Yang C, Wang YJ, Jiang SL, Xu XF, Li XR. Scientometric analysis and historical review of diabetic encephalopathy research: Trends and hotspots (2004-2023). World J Diabetes 2025; 16:91200. [DOI: 10.4239/wjd.v16.i5.91200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 12/18/2024] [Accepted: 02/20/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Diabetic encephalopathy (DE) is a common and serious complication of diabetes that can cause death in many patients and significantly affects the lives of individuals and society. Multiple studies investigating the pathogenesis of DE have been reported. However, few studies have focused on scientometric analysis of DE.
AIM To analyze literature on DE using scientometrics to provide a comprehensive picture of research directions and progress in this field.
METHODS We reviewed studies on DE or cognitive impairment published between 2004 and 2023. The latter were used to identify the most frequent keywords in the keyword analysis and explore the hotspots and trends of DE.
RESULTS Scientometric analysis revealed 1308 research papers on DE, a number that increased annually over the past 20 years, and that the primary topics explored were domain distribution, knowledge structure, evolution, and emergence of research topics related to DE. The inducing factors, comorbidities, pathogenesis, treatment, and animal models of DE help clarify its occurrence, development, and treatment. An increasing number of studies on DE may be a result of the recent increase in patients with diabetes, unhealthy lifestyles, and unhealthy eating habits, which have aggravated the incidence of this disease.
CONCLUSION We identified the main inducing factors and comorbidities of DE, though other complex factors undoubtedly increase social and economic burdens. These findings provide vital references for future studies.
Collapse
Affiliation(s)
- Xian-Wen Ye
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Hai-Xia Zhang
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Qian Li
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Chun-Shuai Li
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Chong-Jun Zhao
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Liang-Jing Xia
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Hong-Min Ren
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xu-Xing Wang
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Chao Yang
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yu-Jie Wang
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shui-Lan Jiang
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xin-Fang Xu
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiang-Ri Li
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| |
Collapse
|
2
|
Zhang S, Shi X, Zheng S, Liang X, Wang F, Xu W, Yu X, Yang Y. The Diabetic Cognitive Impairment Score for Early Screening of Cognitive Impairment in Type 2 Diabetes Patients. J Diabetes Res 2025; 2025:8029913. [PMID: 40271536 PMCID: PMC12017955 DOI: 10.1155/jdr/8029913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/21/2025] [Accepted: 04/04/2025] [Indexed: 04/25/2025] Open
Abstract
Purpose: Diabetes has been associated with an excess risk of cognitive impairment. The hyperphosphorylation of tau protein leads to neurodegeneration and is closely related to Type 2 diabetes (T2D). This study aimed to characterize the association between P-tau181 and diabetic cognitive impairment and to develop a nomogram-based score to screen cognitive impairment in T2D patients. Methods: We used a cohort of 379 patients diagnosed with T2D as a training dataset to develop a predictive model. Risk factors associated with cognitive impairment were identified using stepwise multivariate logistic regressive analysis. A nomogram was established by incorporating these risk factors, and the diabetic cognitive impairment score (DCIS) was built and externally validated in another cohort. Results: In the training cohort, patients with cognitive impairment had higher levels of P-tau181 (13.3 [10.5-18.7] vs. 10.0 [8.0-13.0], p < 0.001). P-tau181 was negatively correlated with MOCA (r = -0.308, p < 0.001) and MMSE (r = -0.289, p < 0.001), and it was independently associated with cognitive impairment in T2D patients (OR, 1.137 [95% CI, 1.080-1.198]; p < 0.001). Other independent risk factors of diabetic cognitive impairment included age, education level, and diabetic retinopathy. The DCIS was built by nomogram based on the four risk factors, which had an area under the receiver operating characteristic curve (AUC) of 0.795 (95% CI, 0.751-0.840). The optimal cut-off of DCIS for the diagnosis of cognitive impairment in T2D patients was 139.5, with a sensitivity of 72.9% and a specificity of 75.3%. In the validation cohort, the AUC of DCIS for screening diabetic cognitive impairment was 0.770 (95% CI, 0.716-0.824). Conclusions: P-tau181 was independently associated with diabetic cognitive impairment. The DCIS, based on P-tau181, age, education level, and diabetic retinopathy, is effective to identify cognitive impairment in T2D patients.
Collapse
Affiliation(s)
- Shujun Zhang
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Hubei Clinical Medical Research Center for Endocrinology and Metabolic Diseases, Wuhan, Hubei Province, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei Province, China
| | - Xiaoli Shi
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Hubei Clinical Medical Research Center for Endocrinology and Metabolic Diseases, Wuhan, Hubei Province, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei Province, China
| | - Shaolin Zheng
- Division of Endocrinology, Jingzhou Hospital Traditional Chinese Medicine, Jingzhou, Hubei Province, China
| | - Xiaoli Liang
- Division of Endocrinology, Wenchang People's Hospital, Wenchang, Hainan Province, China
| | - Fen Wang
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Hubei Clinical Medical Research Center for Endocrinology and Metabolic Diseases, Wuhan, Hubei Province, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei Province, China
| | - Weijie Xu
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Hubei Clinical Medical Research Center for Endocrinology and Metabolic Diseases, Wuhan, Hubei Province, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei Province, China
| | - Xuefeng Yu
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Hubei Clinical Medical Research Center for Endocrinology and Metabolic Diseases, Wuhan, Hubei Province, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei Province, China
| | - Yan Yang
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Hubei Clinical Medical Research Center for Endocrinology and Metabolic Diseases, Wuhan, Hubei Province, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei Province, China
| |
Collapse
|
3
|
Yu X, He H, Wen J, Xu X, Ruan Z, Hu R, Wang F, Ju H. Diabetes-related cognitive impairment: Mechanisms, symptoms, and treatments. Open Med (Wars) 2025; 20:20241091. [PMID: 39822993 PMCID: PMC11737369 DOI: 10.1515/med-2024-1091] [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: 05/11/2024] [Revised: 08/23/2024] [Accepted: 10/18/2024] [Indexed: 01/19/2025] Open
Abstract
Background Diabetes-related cognitive impairment is increasingly recognized as a significant complication, profoundly impacting patients' quality of life. This review aims to examine the pathophysiological mechanisms, clinical manifestations, risk factors, assessment and diagnosis, management strategies, and future research directions of cognitive impairment in diabetes. Methodology A comprehensive literature search was conducted using PubMed, Medline, and other medical databases to identify, review, and evaluate published articles on cognitive impairment in diabetes. The search focused on studies examining pathophysiology, clinical presentations, risk factors, diagnostic approaches, and management strategies. Results The review of current literature revealed that chronic hyperglycemia, insulin resistance, and vascular factors are major contributing factors to cognitive deficits in diabetes. Clinical manifestations include impairments in attention, memory, executive function, visuospatial abilities, and language. Risk factors encompass disease duration, glycemic control, presence of complications, age, education level, and comorbidities. Assessment tools include cognitive screening instruments, neuropsychological testing, and neuroimaging techniques. Management strategies involve glycemic control optimization, lifestyle modifications, cognitive training, and pharmacological interventions. Conclusion This review highlights the significant prevalence and impact of cognitive impairment in diabetes, resulting from complex metabolic and vascular disturbances. Early detection and multifaceted interventions are crucial for preserving cognitive function and improving patient outcomes. Future research should focus on neuroprotective strategies, biomarker identification, and personalized approaches. Collaborative efforts between clinicians and researchers are essential to effectively address this growing healthcare challenge and enhance the quality of life for individuals with diabetes-related cognitive impairment.
Collapse
Affiliation(s)
- Xueting Yu
- Endocrine Department, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, 650000, Yunnan, China
| | - Huimei He
- Endocrine Department, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, 650000, Yunnan, China
| | - Jie Wen
- Executive Ward Department, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, 650000, Yunnan, China
| | - Xiuyuan Xu
- Endocrine Department, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, 650000, Yunnan, China
| | - Zhaojuan Ruan
- Endocrine Department, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, 650000, Yunnan, China
| | - Rui Hu
- Department of Hematology, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650000, Yunnan, China
| | - Fang Wang
- Executive Ward Department, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, 650000, Yunnan, China
| | - Haibing Ju
- Endocrine Department, 920th Hospital of Joint Logistics Support Force, PLA, No. 212 Daguan Road, Xishan District, Kunming, 650000, Yunnan, China
| |
Collapse
|
4
|
Liu S, Chen H, He XD, Yang XO. Glucometabolic-Related Genes as Diagnostic Biomarkers and Therapeutic Targets for Alzheimer's Disease and Type 2 Diabetes Mellitus: A Bioinformatics Analysis. Neurol Res Int 2024; 2024:5200222. [PMID: 38595695 PMCID: PMC11003797 DOI: 10.1155/2024/5200222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 01/26/2024] [Accepted: 02/24/2024] [Indexed: 04/11/2024] Open
Abstract
Background Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM) are two widespread chronic disorders characterized by shared risk factors and molecular pathways. Glucose metabolism, pivotal for cellular homeostasis and energy supply, plays a critical role in these diseases. Its disturbance has been linked to the pathogenesis of both AD and T2DM. However, a comprehensive investigation into the specific roles of glucometabolic genes in the onset and progression of AD and T2DM has yet to be conducted. Methods By analyzing microarray datasets from the Gene Expression Omnibus (GEO) repository, we identified differentially expressed glucometabolic genes (DEGs) in AD and T2DM cohorts. A range of bioinformatics tools were employed for functional annotation, pathway enrichment, protein interaction network construction, module analysis, ROC curve assessment, correlation matrix construction, gene set enrichment analysis, and gene-drug interaction mapping of these DEGs. Key genes were further validated using quantitative real-time polymerase chain reaction (qRT-PCR) in AD and T2DM murine models. Results Our investigation identified 41 glucometabolic-related DEGs, with six prominent genes (G6PD, PKM, ENO3, PFKL, PGD, and TALDO1) being common in both AD and T2DM cohorts. These genes play crucial roles in metabolic pathways including glycolysis, pentose phosphate pathway, and amino sugar metabolism. Their diagnostic potential was highlighted by area under curve (AUC) values exceeding 0.6 for AD and 0.8 for T2DM. Further analysis explored the interactions, pathway enrichments, regulatory mechanisms, and potential drug interactions of these key genes. In the AD murine model, quantitative real-time polymerase chain reaction (qRT-PCR) analysis revealed significant upregulation of G6pd, Eno3, and Taldo1. Similarly, in the T2DM murine model, elevated expression levels of G6pd, Pfkl, Eno3, and Pgd were observed. Conclusion Our rigorous research sheds light on the molecular interconnections between AD and T2DM from a glucometabolic perspective, revealing new opportunities for pharmacological innovation and therapeutic approaches. This study appears to be the first to extensively investigate glucometabolic-associated DEGs and key genes in both AD and T2DM, utilizing multiple datasets. These insights are set to enhance our understanding of the complex pathophysiology underlying these widespread chronic diseases.
Collapse
Affiliation(s)
- Shuo Liu
- The Fourth People's Hospital of Shenyang, Shenyang, Liaoning Province, China
| | - He Chen
- The Fourth People's Hospital of Shenyang, Shenyang, Liaoning Province, China
| | - Xiao-Dong He
- The Fourth People's Hospital of Shenyang, Shenyang, Liaoning Province, China
| | - Xiao-Ou Yang
- The Fourth People's Hospital of Shenyang, Shenyang, Liaoning Province, China
| |
Collapse
|
5
|
Pereira VM, Pradhanang S, Prather JF, Nair S. Role of Metalloproteinases in Diabetes-associated Mild Cognitive Impairment. Curr Neuropharmacol 2024; 23:58-74. [PMID: 38963109 PMCID: PMC11519823 DOI: 10.2174/1570159x22666240517090855] [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: 11/16/2023] [Revised: 01/24/2024] [Accepted: 02/14/2024] [Indexed: 07/05/2024] Open
Abstract
Diabetes has been linked to an increased risk of mild cognitive impairment (MCI), a condition characterized by a subtle cognitive decline that may precede the development of dementia. The underlying mechanisms connecting diabetes and MCI involve complex interactions between metabolic dysregulation, inflammation, and neurodegeneration. A critical mechanism implicated in diabetes and MCI is the activation of inflammatory pathways. Chronic low-grade inflammation, as observed in diabetes, can lead to the production of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), interleukin-1 beta (IL-1β), and interferon-gamma (IFNγ), each of which can exacerbate neuroinflammation and contribute to cognitive decline. A crucial enzyme involved in regulating inflammation is ADAM17, a disintegrin, and metalloproteinase, which can cleave and release TNF-α from its membrane-bound precursor and cause it to become activated. These processes, in turn, activate additional inflammation-related pathways, such as AKT, NF-κB, NLP3, MAPK, and JAK-STAT pathways. Recent research has provided novel insights into the role of ADAM17 in diabetes and neurodegenerative diseases. ADAM17 is upregulated in both diabetes and Alzheimer's disease, suggesting a shared mechanism and implicating inflammation as a possible contributor to much broader forms of pathology and pointing to a possible link between inflammation and the emergence of MCI. This review provides an overview of the different roles of ADAM17 in diabetes-associated mild cognitive impairment diseases. It identifies mechanistic connections through which ADAM17 and associated pathways may influence the emergence of mild cognitive impairment.
Collapse
Affiliation(s)
- Vitoria Mattos Pereira
- School of Pharmacy, College of Health Sciences, Biomedical Sciences, Interdisciplinary Graduate Program, University of Wyoming, Laramie, WY 82071, USA
| | - Suyasha Pradhanang
- School of Pharmacy, College of Health Sciences, Biomedical Sciences, Interdisciplinary Graduate Program, University of Wyoming, Laramie, WY 82071, USA
| | - Jonathan F. Prather
- Department of Zoology and Physiology, Program in Neuroscience, University of Wyoming, Laramie, WY 82071, USA
| | - Sreejayan Nair
- School of Pharmacy, College of Health Sciences, Biomedical Sciences, Interdisciplinary Graduate Program, University of Wyoming, Laramie, WY 82071, USA
| |
Collapse
|
6
|
García-Casares N, González-González G, de la Cruz-Cosme C, Garzón-Maldonado FJ, de Rojas-Leal C, Ariza MJ, Narváez M, Barbancho MÁ, García-Arnés JA, Tinahones FJ. Effects of GLP-1 receptor agonists on neurological complications of diabetes. Rev Endocr Metab Disord 2023; 24:655-672. [PMID: 37231200 PMCID: PMC10404567 DOI: 10.1007/s11154-023-09807-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/21/2023] [Indexed: 05/27/2023]
Abstract
Emerging evidence suggests that treatment with glucagon-like peptide-1 receptor agonists (GLP-1 RAs) could be an interesting treatment strategy to reduce neurological complications such as stroke, cognitive impairment, and peripheral neuropathy. We performed a systematic review to examine the evidence concerning the effects of GLP-1 RAs on neurological complications of diabetes. The databases used were Pubmed, Scopus and Cochrane. We selected clinical trials which analysed the effect of GLP-1 RAs on stroke, cognitive impairment, and peripheral neuropathy. We found a total of 19 studies: 8 studies include stroke or major cardiovascular events, 7 involve cognitive impairment and 4 include peripheral neuropathy. Semaglutide subcutaneous and dulaglutide reduced stroke cases. Liraglutide, albiglutide, oral semaglutide and efpeglenatide, were not shown to reduce the number of strokes but did reduce major cardiovascular events. Exenatide, dulaglutide and liraglutide improved general cognition but no significant effect on diabetic peripheral neuropathy has been reported with GLP-1 RAs. GLP-1 RAs are promising drugs that seem to be useful in the reduction of some neurological complications of diabetes. However, more studies are needed.
Collapse
Affiliation(s)
- Natalia García-Casares
- Facultad de Medicina, Departamento de Medicina, Universidad de Málaga, Málaga, Spain.
- Instituto de Investigación Biomédica de Málaga (I.B.I.M.A), Málaga, Spain.
- Centro de Investigaciones Médico-Sanitarias (C.I.M.ES), Málaga, Spain.
- Departamento de Medicina, Facultad de Medicina, Universidad de Málaga, Centro de Investigaciones Médico Sanitarias (C.I.M.E.S), Universidad de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Campus Universitario de Teatinos s/n., Málaga, 29010, España.
| | | | - Carlos de la Cruz-Cosme
- Instituto de Investigación Biomédica de Málaga (I.B.I.M.A), Málaga, Spain
- Hospital Universitario Virgen de la Victoria de Málaga, Málaga, Spain
| | - Francisco J Garzón-Maldonado
- Instituto de Investigación Biomédica de Málaga (I.B.I.M.A), Málaga, Spain
- Hospital Universitario Virgen de la Victoria de Málaga, Málaga, Spain
| | - Carmen de Rojas-Leal
- Facultad de Medicina, Departamento de Medicina, Universidad de Málaga, Málaga, Spain
| | - María J Ariza
- Facultad de Medicina, Departamento de Medicina, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (I.B.I.M.A), Málaga, Spain
- Centro de Investigaciones Médico-Sanitarias (C.I.M.ES), Málaga, Spain
| | - Manuel Narváez
- Facultad de Medicina, Departamento de Medicina, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (I.B.I.M.A), Málaga, Spain
| | - Miguel Ángel Barbancho
- Facultad de Medicina, Departamento de Medicina, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga (I.B.I.M.A), Málaga, Spain
- Centro de Investigaciones Médico-Sanitarias (C.I.M.ES), Málaga, Spain
| | | | - Francisco J Tinahones
- Facultad de Medicina, Departamento de Medicina, Universidad de Málaga, Málaga, Spain.
- Instituto de Investigación Biomédica de Málaga (I.B.I.M.A), Málaga, Spain.
- Hospital Universitario Virgen de la Victoria de Málaga, Málaga, Spain.
- Departamento de Medicina, Facultad de Medicina, Universidad de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Campus Universitario de Teatinos s/n., Málaga, 29010, España.
| |
Collapse
|
7
|
Goodarzi G, Tehrani SS, Fana SE, Moradi-Sardareh H, Panahi G, Maniati M, Meshkani R. Crosstalk between Alzheimer's disease and diabetes: a focus on anti-diabetic drugs. Metab Brain Dis 2023; 38:1769-1800. [PMID: 37335453 DOI: 10.1007/s11011-023-01225-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 04/26/2023] [Indexed: 06/21/2023]
Abstract
Alzheimer's disease (AD) and Type 2 diabetes mellitus (T2DM) are two of the most common age-related diseases. There is accumulating evidence of an overlap in the pathophysiological mechanisms of these two diseases. Studies have demonstrated insulin pathway alternation may interact with amyloid-β protein deposition and tau protein phosphorylation, two essential factors in AD. So attention to the use of anti-diabetic drugs in AD treatment has increased in recent years. In vitro, in vivo, and clinical studies have evaluated possible neuroprotective effects of anti-diabetic different medicines in AD, with some promising results. Here we review the evidence on the therapeutic potential of insulin, metformin, Glucagon-like peptide-1 receptor agonist (GLP1R), thiazolidinediones (TZDs), Dipeptidyl Peptidase IV (DPP IV) Inhibitors, Sulfonylureas, Sodium-glucose Cotransporter-2 (SGLT2) Inhibitors, Alpha-glucosidase inhibitors, and Amylin analog against AD. Given that many questions remain unanswered, further studies are required to confirm the positive effects of anti-diabetic drugs in AD treatment. So to date, no particular anti-diabetic drugs can be recommended to treat AD.
Collapse
Affiliation(s)
- Golnaz Goodarzi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Student Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pathobiology and Laboratory Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Sadra Samavarchi Tehrani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Student Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Ebrahimi Fana
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Student Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ghodratollah Panahi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmood Maniati
- English Department, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Yin Q, Gao Y, Wang X, Li S, Hou X, Bi W. China should emphasize understanding and standardized management in diabetic cognitive dysfunction. Front Endocrinol (Lausanne) 2023; 14:1195962. [PMID: 37415663 PMCID: PMC10321298 DOI: 10.3389/fendo.2023.1195962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/29/2023] [Indexed: 07/08/2023] Open
Affiliation(s)
- Qingqing Yin
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yan Gao
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xinyu Wang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shangbin Li
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xunyao Hou
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Wenkai Bi
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
9
|
Chen HA, Hovens IB, Davis XS, Hutelin Z, Wall KM, Small DM. Identification of a novel link between adiposity and visuospatial perception. Obesity (Silver Spring) 2023; 31:423-433. [PMID: 36546337 PMCID: PMC9877146 DOI: 10.1002/oby.23603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/19/2022] [Accepted: 09/13/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Recent work has reported a negative association between BMI and performance on the Penn Line Orientation Task. To determine the reliability of this effect, a comprehensive assessment of visual function in individuals with healthy weight (HW) and those with overweight/obesity (OW/OB) was performed. METHODS Visual acuity/contrast, Penn Line Orientation Task, and higher-order visuospatial function were measured in 80 (40 with HW, 40 with OW/OB) case-control study participants. Adiposity, fasting glucose, hemoglobin A1c, diet, physical activity, and heart rate variability were also assessed. A subgroup of 22 participants plus 5 additional participants (n = 27) underwent functional magnetic resonance imaging scanning. RESULTS Compared with those with HW, individuals with OW/OB performed worse on tasks requiring judgments of line orientation. This effect was mediated by body fat percentage and was unrelated to other measures. Functional magnetic resonance imaging revealed a negative association between BMI and response in the primary visual cortex (V1) during line orientation judgment. Performance was unrelated to V1 response but positively correlated with response in a network of regions, including the lateral occipital cortex, when BMI was accounted for in the model. CONCLUSIONS These results demonstrate a selective deficit in line orientation perception associated with adiposity and blunted activation in the V1 that cannot be attributed to visual acuity and does not generalize to other visuospatial tasks.
Collapse
Affiliation(s)
- H. Alexander Chen
- Department of PsychiatryYale University School of Medicine, Yale UniversityNew HavenConnecticutUSA
- Modern Diet and Physiology Research CenterNew HavenConnecticutUSA
| | - Iris B. Hovens
- Department of PsychiatryYale University School of Medicine, Yale UniversityNew HavenConnecticutUSA
- Modern Diet and Physiology Research CenterNew HavenConnecticutUSA
| | - Xue S. Davis
- Department of PsychiatryYale University School of Medicine, Yale UniversityNew HavenConnecticutUSA
- Modern Diet and Physiology Research CenterNew HavenConnecticutUSA
| | - Zach Hutelin
- Department of PsychiatryYale University School of Medicine, Yale UniversityNew HavenConnecticutUSA
- Modern Diet and Physiology Research CenterNew HavenConnecticutUSA
| | - Kathryn M. Wall
- Department of PsychiatryYale University School of Medicine, Yale UniversityNew HavenConnecticutUSA
- Modern Diet and Physiology Research CenterNew HavenConnecticutUSA
| | - Dana M. Small
- Department of PsychiatryYale University School of Medicine, Yale UniversityNew HavenConnecticutUSA
- Modern Diet and Physiology Research CenterNew HavenConnecticutUSA
- Department of PsychologyYale UniversityNew HavenConnecticutUSA
| |
Collapse
|
10
|
Zhang X, Zhu X, Bi X, Huang J, Zhou L. The Insulin Receptor: An Important Target for the Development of Novel Medicines and Pesticides. Int J Mol Sci 2022; 23:7793. [PMID: 35887136 PMCID: PMC9325136 DOI: 10.3390/ijms23147793] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 02/04/2023] Open
Abstract
The insulin receptor (IR) is a transmembrane protein that is activated by ligands in insulin signaling pathways. The IR has been considered as a novel therapeutic target for clinical intervention, considering the overexpression of its protein and A-isoform in multiple cancers, Alzheimer's disease, and Type 2 diabetes mellitus in humans. Meanwhile, it may also serve as a potential target in pest management due to its multiple physiological influences in insects. In this review, we provide an overview of the structural and molecular biology of the IR, functions of IRs in humans and insects, physiological and nonpeptide small molecule modulators of the IR, and the regulating mechanisms of the IR. Xenobiotic compounds and the corresponding insecticidal chemicals functioning on the IR are also discussed. This review is expected to provide useful information for a better understanding of human IR-related diseases, as well as to facilitate the development of novel small-molecule activators and inhibitors of the IR for use as medicines or pesticides.
Collapse
Affiliation(s)
| | | | | | - Jiguang Huang
- Key Laboratory of Natural Pesticides & Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou 510642, China; (X.Z.); (X.Z.); (X.B.)
| | - Lijuan Zhou
- Key Laboratory of Natural Pesticides & Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou 510642, China; (X.Z.); (X.Z.); (X.B.)
| |
Collapse
|
11
|
Vegas-Suárez S, Simón J, Martínez-Chantar ML, Moratalla R. Metabolic Diffusion in Neuropathologies: The Relevance of Brain-Liver Axis. Front Physiol 2022; 13:864263. [PMID: 35634148 PMCID: PMC9134112 DOI: 10.3389/fphys.2022.864263] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/25/2022] [Indexed: 11/10/2022] Open
Abstract
Chronic liver diseases include a broad group of hepatic disorders from different etiologies and with varying degrees of progression and severity. Among them, non-alcoholic fatty (NAFLD) and alcoholic (ALD) liver diseases are the most frequent forms of expression, caused by either metabolic alterations or chronic alcohol consumption. The liver is the main regulator of energy homeostasis and metabolism of potentially toxic compounds in the organism, thus hepatic disorders often promote the release of harmful substances. In this context, there is an existing interconnection between liver and brain, with the well-named brain-liver axis, in which liver pathologies lead to the promotion of neurodegenerative disorders. Alzheimer's (AD) and Parkinson's (PD) diseases are the most relevant neurological disorders worldwide. The present work highlights the relevance of the liver-related promotion of these disorders. Liver-related hyperammonemia has been related to the promotion of perturbations in nervous systems, whereas the production of ketone bodies under certain conditions may protect from developing them. The capacity of the liver of amyloid-β (Aβ) clearance is reduced under liver pathologies, contributing to the development of AD. These perturbations are even aggravated by the pro-inflammatory state that often accompanies liver diseases, leading to the named neuroinflammation. The current nourishment habits, named as Western diet (WD) and alterations in the bile acid (BA) profile, whose homeostasis is controlled by the liver, have been also related to both AD and PD, whereas the supplementation with certain compounds, has been demonstrated to alleviate the pathologies.
Collapse
Affiliation(s)
- Sergio Vegas-Suárez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
- Cajal Institute, Spanish National Research Council (CSIC), Madrid, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERned), Carlos III Institute of Health (ISCIII), Madrid, Spain
| | - Jorge Simón
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III Institute of Health (ISCIII), Madrid, Spain
| | - María Luz Martínez-Chantar
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III Institute of Health (ISCIII), Madrid, Spain
| | - Rosario Moratalla
- Cajal Institute, Spanish National Research Council (CSIC), Madrid, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERned), Carlos III Institute of Health (ISCIII), Madrid, Spain
| |
Collapse
|
12
|
Angoff R, Himali JJ, Maillard P, Aparicio HJ, Vasan RS, Seshadri S, Beiser AS, Tsao CW. Relations of Metabolic Health and Obesity to Brain Aging in Young to Middle-Aged Adults. J Am Heart Assoc 2022; 11:e022107. [PMID: 35229662 PMCID: PMC9075324 DOI: 10.1161/jaha.121.022107] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 01/18/2022] [Indexed: 11/23/2022]
Abstract
Background We aimed to evaluate the association between metabolic health and obesity and brain health measured via magnetic resonance imaging and neurocognitive testing in community dwelling adults. Methods and Results Framingham Heart Study Third Generation Cohort members (n=2170, 46±9 years of age, 54% women) without prevalent diabetes, stroke, dementia, or other neurologic conditions were grouped by metabolic unhealthiness (≥2 criteria for metabolic syndrome) and obesity (body mass index ≥30 kg/m2): metabolically healthy (MH) nonobese, MH obese, metabolically unhealthy (MU) nonobese, and MU obese. We evaluated the relationships of these groups with brain structure (magnetic resonance imaging) and function (neurocognitive tests). In multivariable-adjusted analyses, metabolically unhealthy individuals (MU nonobese and MU obese) had lower total cerebral brain volume compared with the MH nonobese referent group (both P<0.05). Additionally, the MU obese group had greater white matter hyperintensity volume (P=0.004), whereas no association was noted between white matter hyperintensity volume and either groups of metabolic health or obesity alone. Obese individuals had less favorable cognitive scores: MH obese had lower scores on global cognition, Logical Memory-Delayed Recall and Similarities tests, and MU obese had lower scores on Similarities and Visual Reproductions-Delayed tests (all P≤0.04). MU and obese groups had higher free water content and lower fractional anisotropy in several brain regions, consistent with loss of white matter integrity. Conclusions In this cross-sectional cohort study of younger to middle-aged adults, poor metabolic health and obesity were associated with structural and functional evidence of brain aging. Improvement in metabolic health and obesity may present opportunities to improve long-term brain health.
Collapse
Affiliation(s)
- Rebecca Angoff
- Cardiovascular DivisionBeth Israel Deaconess Medical Center and Harvard Medical SchoolBostonMA
| | - Jayandra J. Himali
- Department of NeurologySchool of MedicineBoston UniversityBostonMA
- The Department of BiostatisticsBoston UniversityBostonMA
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative DiseasesUniversity of Texas Health Sciences CenterSan AntonioTX
- The Framingham Heart StudyFraminghamMA
| | - Pauline Maillard
- Department of Neurology and Center for NeuroscienceUniversity of California at DavisDavisCA
| | - Hugo J. Aparicio
- Department of NeurologySchool of MedicineBoston UniversityBostonMA
- The Framingham Heart StudyFraminghamMA
| | - Ramachandran S. Vasan
- Department of MedicineSchool of MedicineBoston UniversityBostonMA
- Department of EpidemiologyBoston UniversityBostonMA
- The Framingham Heart StudyFraminghamMA
| | - Sudha Seshadri
- Department of NeurologySchool of MedicineBoston UniversityBostonMA
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative DiseasesUniversity of Texas Health Sciences CenterSan AntonioTX
- Department of Population Health SciencesUniversity of Texas Health Science CenterSan AntonioTX
- The Framingham Heart StudyFraminghamMA
| | - Alexa S. Beiser
- Department of NeurologySchool of MedicineBoston UniversityBostonMA
- The Department of BiostatisticsBoston UniversityBostonMA
- The Framingham Heart StudyFraminghamMA
| | - Connie W. Tsao
- Cardiovascular DivisionBeth Israel Deaconess Medical Center and Harvard Medical SchoolBostonMA
- The Framingham Heart StudyFraminghamMA
| |
Collapse
|
13
|
Neuroimaging and modulation in obesity and diabetes research: 10th anniversary meeting. Int J Obes (Lond) 2022; 46:718-725. [PMID: 34934178 PMCID: PMC8960390 DOI: 10.1038/s41366-021-01025-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 10/29/2021] [Accepted: 11/09/2021] [Indexed: 11/29/2022]
|
14
|
Alshaikh AA, Alatawi HS, Alzahrani AM. Neurocognitive dysfunction among type 2 diabetes patients attending primary health care in Jeddah, Saudi Arabia. J Family Med Prim Care 2021; 10:3803-3814. [PMID: 34934684 PMCID: PMC8653469 DOI: 10.4103/jfmpc.jfmpc_423_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/05/2021] [Accepted: 07/19/2021] [Indexed: 11/04/2022] Open
Abstract
CONTEXT Identification of modifiable risk factors of neurocognitive dysfunction (NCDs) that would help in preventing neurocognitive dysfunction by means of appropriate measures. OBJECTIVES The study aims to provide an insight into the extent and cofactors ofNCDs among Saudi type II diabetes (T2DM) patients at the primary care level. SETTINGS AND DESIGN A cross-sectional study was conducted at five randomly selected primary health care centers (PHCCs) of Jeddah, Saudi Arabia. SUBJECT AND METHODS T2DM patients above 40 years, who were followed for T2DM diagnosed ≥ 1 year ago were recruited. The Arabic version of the Mini-Mental State Evaluation (MMSE) was used for screening NCDs, using education-adjusted cut-offs. STATISTICAL ANALYSIS USED Sociodemographic, diabetes-related, and other clinical and lifestyle factors were analyzed as cofactors of NCDs. RESULTS The study included 236 T2DM patients, who had mean ± SD age of 60.29 ± 9.45 years. The majority (61.0%) were female, and mean ± SD duration of T2DM was 14.1 ± 8.4 years (range = 1-45 years). The prevalence of NCDs was 35.2% (95% CI = 29.1%, 41.6%), and 5.1% of the participants had MMSE scores ≤ 10 indicating severe neurocognitive impairment. The congruence of significant sociodemographic factors delineated a high-risk profile, and multivariate regression analysis showed female gender, low educational level, longer duration of diabetes, geriatric age at T2DM diagnosis, inadequate glycemic control, and sedentary lifestyle as the independent risk factors for NCDs. CONCLUSIONS The population of middle-aged and older T2DM patients is highly exposed to NCDs, with the great contribution of other comorbidities and higher risk incurred by older, lowly educated females with long diabetes duration. Further improvements should be achieved to enhance the care offered to diabetic patients by improving glycemic control, screening for comorbidities, and early detection of neurocognitive decline.
Collapse
Affiliation(s)
- Alshaymaa A Alshaikh
- Department of Family Medicine, King Abdulaziz Medical City, Jeddah, Saudi Arabia
| | - Hind S Alatawi
- Department of Family Medicine, King Fahad Armed Forces Hospital, Jeddah, Saudi Arabia
| | - Abdullah M Alzahrani
- Department of Family Medicine, King Saud Bin Abdulaziz University for Health and Sciences, King Abdulaziz Medical City, King Abdullah Inteernational Medical Research Center, Jeddah, Saudi Arabia
| |
Collapse
|
15
|
The "Adipo-Cerebral" Dialogue in Childhood Obesity: Focus on Growth and Puberty. Physiopathological and Nutritional Aspects. Nutrients 2021; 13:nu13103434. [PMID: 34684432 PMCID: PMC8539184 DOI: 10.3390/nu13103434] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 01/08/2023] Open
Abstract
Overweight and obesity in children and adolescents are overwhelming problems in western countries. Adipocytes, far from being only fat deposits, are capable of endocrine functions, and the endocrine activity of adipose tissue, resumable in adipokines production, seems to be a key modulator of central nervous system function, suggesting the existence of an “adipo-cerebral axis.” This connection exerts a key role in children growth and puberty development, and it is exemplified by the leptin–kisspeptin interaction. The aim of this review was to describe recent advances in the knowledge of adipose tissue endocrine functions and their relations with nutrition and growth. The peculiarities of major adipokines are briefly summarized in the first paragraph; leptin and its interaction with kisspeptin are focused on in the second paragraph; the third paragraph deals with the regulation of the GH-IGF axis, with a special focus on the model represented by growth hormone deficiency (GHD); finally, old and new nutritional aspects are described in the last paragraph.
Collapse
|
16
|
Liraglutide Alleviates Cognitive Deficit in db/db Mice: Involvement in Oxidative Stress, Iron Overload, and Ferroptosis. Neurochem Res 2021; 47:279-294. [PMID: 34480710 DOI: 10.1007/s11064-021-03442-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/28/2021] [Accepted: 08/30/2021] [Indexed: 12/11/2022]
Abstract
Studies have shown that diabetes is associated with the occurrence of neurodegenerative diseases and cognitive decline. However, there is currently no effective treatment for diabetes-induced cognitive dysfunction. The superior efficacy of liraglutide (LIRA) for cognitive impairment and numerous neurodegenerative diseases has been widely demonstrated. This study determined the effects of LIRA on diabetic cognitive impairment and on the levels of oxidative stress, lipid peroxidation, iron metabolism and ferroptosis in the hippocampus. Mice were injected daily with liraglutide (200 μg/kg/d) for 5 weeks. LIRA could repair damaged neurons and synapses, and it increased the protein expression levels of PSD 95, SYN, and BDNF. Furthermore, LIRA significantly decreased oxidative stress and lipid peroxidation levels by downregulating the production of ROS and MDA and upregulating SOD and GSH-Px in the serum and hippocampus, and the upregulation of SOD2 expression was also proven. The decreased levels of TfR1 and the upregulation of FPN1 and FTH proteins observed in the LIRA-treated db/db group were shown to reduce iron overload in the hippocampus, whereas the increased expression of Mtft and decreased expression of Mfrn in the mitochondria indicated that mitochondrial iron overload was ameliorated. Finally, LIRA was shown to prevent ferroptosis in the hippocampus by elevating the expression of GPX4 and SLC7A11 and suppressing the excessive amount of ACSL4; simultaneously, the damage to the mitochondria observed by TEM was also repaired. For the first time, we proved in the T2DM model that ferroptosis occurs in the hippocampus, which may play a role in diabetic cognitive impairment. LIRA can reduce oxidative stress, lipid peroxidation and iron overload in diabetic cognitive disorders and further inhibit ferroptosis, thereby weakening the damage to hippocampal neurons and synaptic plasticity and ultimately restoring cognitive function.
Collapse
|
17
|
Ryu WI, Cohen BM, Sonntag KC. Hypothesis and Theory: Characterizing Abnormalities of Energy Metabolism Using a Cellular Platform as a Personalized Medicine Approach for Alzheimer's Disease. Front Cell Dev Biol 2021; 9:697578. [PMID: 34395428 PMCID: PMC8363296 DOI: 10.3389/fcell.2021.697578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/27/2021] [Indexed: 01/07/2023] Open
Abstract
Sporadic or late-onset Alzheimer's disease (LOAD) is characterized by slowly progressive deterioration and death of CNS neurons. There are currently no substantially disease-modifying therapies. LOAD pathology is closely related to changes with age and include, among others, accumulation of toxic molecules and altered metabolic, microvascular, biochemical and inflammatory processes. In addition, there is growing evidence that cellular energy deficits play a critical role in aging and LOAD pathophysiology. However, the exact mechanisms and causal relationships are largely unknown. In our studies we tested the hypothesis that altered bioenergetic and metabolic cell functions are key elements in LOAD, using a cellular platform consisting of skin fibroblasts derived from LOAD patients and AD-unaffected control individuals and therefrom generated induced pluripotent stem cells that are differentiated to brain-like cells to study LOAD pathogenic processes in context of age, disease, genetic background, cell development, and cell type. This model has revealed that LOAD cells exhibit a multitude of bioenergetic and metabolic alterations, providing evidence for an innate inefficient cellular energy management in LOAD as a prerequisite for the development of neurodegenerative disease with age. We propose that this cellular platform could ultimately be used as a conceptual basis for a personalized medicine tool to predict altered aging and risk for development of dementia, and to test or implement customized therapeutic or disease-preventive intervention strategies.
Collapse
Affiliation(s)
- Woo-In Ryu
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, United States
- Basic Neuroscience Division, McLean Hospital, Harvard Medical School, Belmont, MA, United States
- Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, MA, United States
| | - Bruce M. Cohen
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, United States
- Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, MA, United States
- Psychotic Disorders Division, McLean Hospital, Harvard Medical School, Belmont, MA, United States
| | - Kai-C. Sonntag
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, United States
- Basic Neuroscience Division, McLean Hospital, Harvard Medical School, Belmont, MA, United States
- Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, MA, United States
| |
Collapse
|
18
|
Intranasal Insulin Administration to Prevent Delayed Neurocognitive Recovery and Postoperative Neurocognitive Disorder: A Narrative Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18052681. [PMID: 33799976 PMCID: PMC7967645 DOI: 10.3390/ijerph18052681] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/26/2021] [Accepted: 03/02/2021] [Indexed: 11/29/2022]
Abstract
Delayed neurocognitive recovery and postoperative neurocognitive disorders are major complications of surgery, hospitalization, and anesthesia that are receiving increasing attention. Their incidence is reported to be 10–80% after cardiac surgery and 10–26% after non-cardiac surgery. Some of the risk factors include advanced age, level of education, history of diabetes mellitus, malnutrition, perioperative hyperglycemia, depth of anesthesia, blood pressure fluctuation during surgery, chronic respiratory diseases, etc. Scientific evidence suggests a causal association between anesthesia and delayed neurocognitive recovery or postoperative neurocognitive disorders, and various pathophysiological mechanisms have been proposed: mitochondrial dysfunction, neuroinflammation, increase in tau protein phosphorylation, accumulation of amyloid-β protein, etc. Insulin receptors in the central nervous system have a non-metabolic role and act through a neuromodulator-like action, while an interaction between anesthetics and central nervous system insulin receptors might contribute to anesthesia-induced delayed neurocognitive recovery or postoperative neurocognitive disorders. Acute or chronic intranasal insulin administration, which has no influence on the blood glucose concentration, appears to improve working memory, verbal fluency, attention, recognition of objects, etc., in animal models, cognitively healthy humans, and memory-impaired patients by restoring the insulin receptor signaling pathway, attenuating anesthesia-induced tau protein hyperphosphorylation, etc. The aim of this review is to report preclinical and clinical evidence of the implication of intranasal insulin for preventing changes in the brain molecular pattern and/or neurobehavioral impairment, which influence anesthesia-induced delayed neurocognitive recovery or postoperative neurocognitive disorders.
Collapse
|
19
|
Wittwer J, Bradley D. Clusterin and Its Role in Insulin Resistance and the Cardiometabolic Syndrome. Front Immunol 2021; 12:612496. [PMID: 33717095 PMCID: PMC7946829 DOI: 10.3389/fimmu.2021.612496] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
The cardiometabolic syndrome involves a clustering of metabolic and cardiovascular factors which increase the risk of patients developing both Type 2 Diabetes Mellitus and cardio/cerebrovascular disease. Although the mechanistic underpinnings of this link remain uncertain, key factors include insulin resistance, excess visceral adiposity, atherogenic dyslipidemia, and endothelial dysfunction. Of these, a state of resistance to insulin action in overweight/obese patients appears to be central to the pathophysiologic process. Given the increasing prevalence of obesity-related Type 2 Diabetes, coupled with the fact that cardiovascular disease is the number one cause of mortality in this patient population, a more thorough understanding of the cardiometabolic syndrome and potential options to mitigate its risk is imperative. Inherent in the pathogenesis of insulin resistance is an underlying state of chronic inflammation, at least partly in response to excess adiposity. Within obese adipose tissue, an immunomodulatory shift occurs, involving a preponderance of pro-inflammatory immune cells and cytokines/adipokines, along with antigen presentation by adipocytes. Therefore, various adipokines differentially expressed by obese adipocytes may have a significant effect on cardiometabolism. Clusterin is a molecular chaperone that is widely produced by many tissues throughout the body, but is also preferentially overexpressed by obese compared lean adipocytes and relates strongly to multiple components of the cardiometabolic syndrome. Herein, we summarize the known and potential roles of circulating and adipocyte-specific clusterin in cardiometabolism and discuss potential further investigations to determine if clusterin is a viable target to attenuate both metabolic and cardiovascular disease.
Collapse
Affiliation(s)
- Jennifer Wittwer
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Diabetes and Metabolism Research Center, The Ohio State University, Columbus, OH, United States
| | - David Bradley
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Diabetes and Metabolism Research Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
20
|
Malan L, Hamer M, von Känel R, van Wyk RD, Sumner AE, Nilsson PM, Lambert GW, Steyn HS, Badenhorst CJ, Malan NT. A Stress Syndrome Prototype Reflects Type 3 Diabetes and Ischemic Stroke Risk: The SABPA Study. BIOLOGY 2021; 10:162. [PMID: 33670473 PMCID: PMC7922484 DOI: 10.3390/biology10020162] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/28/2021] [Accepted: 02/09/2021] [Indexed: 12/19/2022]
Abstract
Type 3 diabetes (T3D) accurately reflects that dementia, e.g., Alzheimer's disease, represents insulin resistance and neurodegeneration in the brain. Similar retinal microvascular changes were observed in Alzheimer's and chronic stressed individuals. Hence, we aimed to show that chronic stress relates to T3D dementia signs and retinopathy, ultimately comprising a Stress syndrome prototype reflecting risk for T3D and stroke. A chronic stress and stroke risk phenotype (Stressed) score, independent of age, race or gender, was applied to stratify participants (N = 264; aged 44 ± 9 years) into high stress risk (Stressed, N = 159) and low stress risk (non-Stressed, N = 105) groups. We determined insulin resistance using the homeostatic model assessment (HOMA-IR), which is interchangeable with T3D, and dementia risk markers (cognitive executive functioning (cognitiveexe-func); telomere length; waist circumference (WC), neuronal glia injury; neuron-specific enolase/NSE, S100B). Retinopathy was determined in the mydriatic eye. The Stressed group had greater incidence of HOMA-IR in the upper quartile (≥5), larger WC, poorer cognitiveexe-func control, shorter telomeres, consistently raised neuronal glia injury, fewer retinal arteries, narrower arteries, wider veins and a larger optic cup/disc ratio (C/D) compared to the non-Stressed group. Furthermore, of the stroke risk markers, arterial narrowing was related to glaucoma risk with a greater C/D, whilst retinal vein widening was related to HOMA-IR, poor cognitiveexe-func control and neuronal glia injury (Adjusted R2 0.30; p ≤ 0.05). These associations were not evident in the non-Stressed group. Logistic regression associations between the Stressed phenotype and four dementia risk markers (cognitiveexe-func, telomere length, NSE and WC) comprised a Stress syndrome prototype (area under the curve 0.80; sensitivity/specificity 85%/58%; p ≤ 0.001). The Stress syndrome prototype reflected risk for HOMA-IR (odds ratio (OR) 7.72) and retinal glia ischemia (OR 1.27) and vein widening (OR 1.03). The Stressed phenotype was associated with neuronal glia injury and retinal ischemia, potentiating glaucoma risk. The detrimental effect of chronic stress exemplified a Stress syndrome prototype reflecting risk for type 3 diabetes, neurodegeneration and ischemic stroke.
Collapse
Affiliation(s)
- Leoné Malan
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom 2520, South Africa; (R.v.K.); (N.T.M.)
| | - Mark Hamer
- Division of Surgery & Interventional Science, Faculty of Medical Sciences, University College London, London WC1E 6BT, UK;
| | - Roland von Känel
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom 2520, South Africa; (R.v.K.); (N.T.M.)
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Roelof D. van Wyk
- Surgical Ophthalmologist, 85 Peter Mokaba Street, Potchefstroom 2531, South Africa;
| | - Anne E. Sumner
- Section on Ethnicity and Health, Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA;
- National Institute of Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peter M. Nilsson
- Department of Clinical Sciences, Lund University, SE-205 02 Malmö, Sweden;
| | - Gavin W. Lambert
- Iverson Health Innovation Research Institute, Swinburne University of Technology, Hawthorn, VIC 3122, Australia;
- Baker Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Hendrik S. Steyn
- Statistical Consultation Services, North-West University, Potchefstroom 2520, South Africa;
| | - Casper J. Badenhorst
- Anglo American Corporate Services, Sustainable Development Department, Johannesburg 2017, South Africa;
| | - Nico T. Malan
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom 2520, South Africa; (R.v.K.); (N.T.M.)
| |
Collapse
|
21
|
Insulin Bidirectionally Alters NAc Glutamatergic Transmission: Interactions between Insulin Receptor Activation, Endogenous Opioids, and Glutamate Release. J Neurosci 2021; 41:2360-2372. [PMID: 33514676 PMCID: PMC7984597 DOI: 10.1523/jneurosci.3216-18.2021] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 12/27/2020] [Accepted: 01/21/2021] [Indexed: 01/11/2023] Open
Abstract
Human fMRI studies show that insulin influences brain activity in regions that mediate reward and motivation, including the nucleus accumbens (NAc). Insulin receptors are expressed by NAc medium spiny neurons (MSNs), and studies of cultured cortical and hippocampal neurons suggest that insulin influences excitatory transmission via presynaptic and postsynaptic mechanisms. However, nothing is known about how insulin influences excitatory transmission in the NAc. Human fMRI studies show that insulin influences brain activity in regions that mediate reward and motivation, including the nucleus accumbens (NAc). Insulin receptors are expressed by NAc medium spiny neurons (MSNs), and studies of cultured cortical and hippocampal neurons suggest that insulin influences excitatory transmission via presynaptic and postsynaptic mechanisms. However, nothing is known about how insulin influences excitatory transmission in the NAc. Furthermore, insulin dysregulation accompanying obesity is linked to cognitive decline, depression, anxiety, and altered motivation that rely on NAc excitatory transmission. Using whole-cell patch-clamp and biochemical approaches, we determined how insulin affects NAc glutamatergic transmission in nonobese and obese male rats and the underlying mechanisms. We find that there are concentration-dependent, bidirectional effects of insulin on excitatory transmission, with insulin receptor activation increasing and IGF receptor activation decreasing NAc excitatory transmission. Increases in excitatory transmission were mediated by activation of postsynaptic insulin receptors located on MSNs. However, this effect was due to an increase in presynaptic glutamate release. This suggested feedback from MSNs to presynaptic terminals. In additional experiments, we found that insulin-induced increases in presynaptic glutamate release are mediated by opioid receptor-dependent disinhibition. Furthermore, obesity resulted in a loss of insulin receptor-mediated increases in excitatory transmission and a reduction in NAc insulin receptor surface expression, while preserving reductions in transmission mediated by IGF receptors. These results provide the first insights into how insulin influences excitatory transmission in the adult brain, and evidence for a previously unidentified form of opioid receptor-dependent disinhibition of NAc glutamatergic transmission. SIGNIFICANCE STATEMENT Data here provide the first insights into how insulin influences excitatory transmission in the adult brain, and identify previously unknown interactions between insulin receptor activation, opioids, and glutamatergic transmission. These data contribute to our fundamental understanding of insulin's influence on brain motivational systems and have implications for the use of insulin as a cognitive enhancer and for targeting of insulin receptors and IGF receptors to alter motivation.
Collapse
|
22
|
Chang Y, Kim CK, Kim MK, Seo WK, Oh K. Insulin resistance is associated with poor functional outcome after acute ischemic stroke in non-diabetic patients. Sci Rep 2021; 11:1229. [PMID: 33441784 PMCID: PMC7806587 DOI: 10.1038/s41598-020-80315-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 11/17/2020] [Indexed: 11/24/2022] Open
Abstract
Insulin resistance is associated with the occurrence of stroke and atherosclerotic disease. However, the relationship between insulin resistance and the prognosis of acute ischemic stroke in non-diabetic patients is unclear. We hypothesized that insulin resistance might affect short-term functional recovery after acute ischemic stroke in non-diabetic patients. Between May 2014 and December 2016, 1377 consecutive patients with acute ischemic stroke were enrolled from a prospectively maintained stroke registry. After excluding patients with transient ischemic attacks (TIA), pre-stroke disabilities, diabetes mellitus, and patients with incomplete evaluations, 517 patients were included in the study. The homeostasis model assessment of insulin resistance (HOMA-IR) score was used to evaluate the degree of insulin resistance. The patients with the highest quartile of log HOMA-IR index scores were younger and had higher fasting blood glucose, total cholesterol, triglycerides, low-density lipoprotein, and HbA1c levels. Multivariable logistic regression analysis revealed that log HOMA-IR scores were independently associated with poor prognosis after adjusting for age and sex and p < 0.1 in univariable analysis. Insulin resistance was associated with the poor functional outcome of non-diabetic stroke patients. This evidence supports treating insulin resistance in acute ischemic stroke patients with blood glucose levels within the normal range.
Collapse
Affiliation(s)
- Yoonkyung Chang
- Department of Neurology, Ewha Womans University Mokdong Hospital and Ewha University College of Medicine, 1071 Anyangcheon-ro, Yangcheon-gu, Seoul, South Korea
| | - Chi Kyung Kim
- Department of Neurology, Korea University Guro Hospital and Korea University College of Medicine, 148 Gurodong-Ro, Guro-gu, Seoul, 08308, South Korea
| | - Min-Kyung Kim
- Department of Neurology, Korea University Guro Hospital and Korea University College of Medicine, 148 Gurodong-Ro, Guro-gu, Seoul, 08308, South Korea
| | - Woo-Keun Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81, Irwon-ro, Gangnam-gu, Seoul, South Korea
| | - Kyungmi Oh
- Department of Neurology, Korea University Guro Hospital and Korea University College of Medicine, 148 Gurodong-Ro, Guro-gu, Seoul, 08308, South Korea.
| |
Collapse
|
23
|
Carmichael OT, Neiberg RH, Dutton GR, Hayden KM, Horton E, Pi-Sunyer FX, Johnson KC, Rapp SR, Spira AP, Espeland MA. Long-term Change in Physiological Markers and Cognitive Performance in Type 2 Diabetes: The Look AHEAD Study. J Clin Endocrinol Metab 2020; 105:5897494. [PMID: 32845968 PMCID: PMC7566388 DOI: 10.1210/clinem/dgaa591] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 12/20/2022]
Abstract
CONTEXT The effects of physiological improvements on cognitive function among persons with type 2 diabetes mellitus (T2DM) are not fully understood. OBJECTIVE To determine whether improvements in physiological markers (body weight, blood sugar control, and physical activity) during intensive lifestyle intervention (ILI) are associated with enhancements in cognitive function in older adults with T2DM. DESIGN Multisite randomized controlled trial. SETTING Academic research centers. PATIENTS OR OTHER PARTICIPANTS Participants were aged 45-76 years, with T2DM. INTERVENTION The Action for Health in Diabetes (Look AHEAD) study, a randomized, controlled clinical trial of ILI. MAIN OUTCOME MEASURE Two to 3 cognitive assessments were collected from 1089 participants, the first and last occurring a mean (standard deviation) of 8.6 (1.0) and 11.5 (0.7) years after enrollment. RESULTS Greater improvement in blood sugar control was associated with better cognitive scores (fasting glucose and Rey Auditory Verbal Learning Test [AVLT]: P = 0.0148; fasting glucose and Digit Symbol Coding (DSC): P = 0.0360; HbA1C and DSC: P = 0.0477); but weight loss had mixed associations with cognitive scores (greater body mass index [BMI] reduction and worse AVLT overall: P = 0.0053; and greater BMI reduction and better DSC scores among those overweight but not obese at baseline: P = 0.010). Associations were strongest among those who were overweight (not obese) at baseline, and among those with a history of cardiovascular disease (CVD) at baseline. CONCLUSIONS Improvements in glycemic control, but not necessarily weight status, during ILI may be associated with better subsequent cognitive performance. These associations may differ by adiposity and CVD history.
Collapse
Affiliation(s)
- Owen T Carmichael
- Biomedical Imaging Center, Pennington Biomedical Research Center, Baton Rouge, Louisiana
- Correspondence and Reprint Requests: Owen T. Carmichael, PhD, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA 70808, USA. E-mail:
| | - Rebecca H Neiberg
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Gareth R Dutton
- Department of Medicine, Division of Preventive Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kathleen M Hayden
- Social Sciences and Health Policy, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Edward Horton
- Joslin Diabetes Center, Harvard University, Boston, Massachusetts
| | - F Xavier Pi-Sunyer
- Division of Endocrinology, Obesity/Nutrition Research Center, Columbia University College of Physicians and Surgeons, New York, NY
| | - Karen C Johnson
- Department of Preventive Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Stephen R Rapp
- Department of Psychiatry & Behavioral Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Adam P Spira
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | - Mark A Espeland
- Division of Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW Research has consistently shown that type 2 diabetes (T2D) is associated with increased risk of all-cause dementia. Because one of the most common clinical presentations of early stage dementia is memory impairment, we examined the relationship of T2D with memory function, using the recently published scientific literature. RECENT FINDINGS We conducted a structured review to identify studies of "T2D and memory" published since 2015. After review of the 129 articles retrieved, we identified 14 studies meeting the inclusion and exclusion criteria. Among the eight studies with a single assessment of memory function in time (mostly cross-sectional), six found an association of T2D with lower memory function, but mostly in select subgroups of persons. Separately, six studies included repeated measures of memory (longitudinal design). Four out of six longitudinal studies found that T2D was related with a faster decline in memory, while two did not. Among the four studies showing a relation with memory decline, two had sample sizes of 9000-10,000 persons. Further, three longitudinal studies controlled for hypertension and stroke as covariates, and results suggested that common vascular risk factors and diseases do not account for the relation. While mechanistic studies clearly support a role for cerebrovascular disease in the relation of T2D with cognition, emerging data suggest that insulin resistance in the brain itself may also play a role. Most, but not all, recently published studies suggest that T2D is associated with a lower level and faster decline in memory function. This association does not appear to be fully accounted for by common vascular processes. More research will clarify the mechanisms linking T2D to memory and dementia.
Collapse
|
25
|
Bradley D. Clusterin as a Potential Biomarker of Obesity-Related Alzheimer's Disease Risk. Biomark Insights 2020; 15:1177271920964108. [PMID: 33110346 PMCID: PMC7555556 DOI: 10.1177/1177271920964108] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 09/14/2020] [Indexed: 02/03/2023] Open
Abstract
Over 35% of the adult US population is obese. In turn, excess adiposity increases the risk of multiple complications including type 2 diabetes (T2D), insulin resistance, and cardiovascular disease; yet, obesity also independently heightens risk of Alzheimer's Disease (AD), even after adjusting for other important confounding risk factors including blood pressure, sociodemographics, cholesterol levels, smoking status, and Apolipoprotein E (ApoE) genotype. Among patients over the age of 65 with dementia, 37% have coexisting diabetes, and an estimated 7.3% of cases of AD are directly attributable to midlife obesity. Clusterin, also known as apolipoprotein J (ApoJ), is a multifunctional glycoprotein that acts as a molecular chaperone, assisting folding of secreted proteins. Clusterin has been implicated in several physiological and pathological states, including AD, metabolic disease, and cardiovascular disease. Despite long-standing interest in elucidating clusterin's relationship with amyloid beta (Aβ) aggregation/clearance and toxicity, significant knowledge gaps still exist. Altered clusterin expression and protein levels have been linked with cognitive and memory function, disrupted central nervous system lipid flux, as well as pathogenic brain structure; and its role in cardiometabolic disease suggests that it may be a link between insulin resistance, dyslipidemia, and AD. Here, we briefly highlight clusterin's relevance to AD by presenting existing evidence linking clusterin to AD and cardiometabolic disease, and discussing its potential utility as a biomarker for AD in the presence of obesity-related metabolic disease.
Collapse
Affiliation(s)
- David Bradley
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
26
|
Sakr HI, Amen MA, Rashed LA, Khowailed AA, Sayed HA, Motawee ME, Sakr H, Khalifa MM. Comparing prophylactic effect of exercise and metformin on cognitive brain functions in rats with type 3 diabetes mellitus. Arch Med Sci 2020; 20:618-631. [PMID: 38757028 PMCID: PMC11094824 DOI: 10.5114/aoms.2020.99023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 03/26/2020] [Indexed: 05/18/2024] Open
Abstract
Introduction Type 2 diabetes mellitus (DM) and Alzheimer's disease (AD) are two major medical conditions that constitute a significant financial burden on most healthcare systems. Due to AD sharing "insulin resistance" mechanistic features with DM, some scientists have proposed "type 3 DM" terminology for it. This study aims to compare the prophylactic effect of exercise and metformin on cognitive brain functions in rats with type 3 DM. Material and methods Two groups of rats were included in the study: the control group (n = 15) and the streptozotocin-induced type 2 diabetic group (n = 45). The diabetic group was subdivided into three equal subgroups: a sedentary non-treated diabetic group, an exercised group, and a metformin-treated group. We estimated step-down avoidance task latency, serum glucose, insulin, free fatty acids (FFA), cholesterol, high-density lipoprotein (HDL), low-density lipoprotein (LDL) and triglycerides (TG), brain Aβ-42 and glucose, histological changes by toluidine blue, and immunohistochemistry for brain Aβ-42 and tau-positive cells. Results Serum glucose, FFA, TG, cholesterol, LDL, brain Aβ-42, brain glucose, the number of hippocampal dark and degenerated cells, and brain Aβ-42 and tau-positive cells, were all significantly lower. In contrast, serum insulin and HDL, the number of hippocampal granular cells, and latency of the step-down avoidance task were significantly higher in exercised and metformin-treated groups compared to the diabetic group. There were significantly higher values of serum insulin and brain/plasma glucose ratio and number of brain tau-positive cells in the metformin-treated group than in the exercised group. Conclusions We can conclude that exercise can be as effective as metformin regarding prophylaxis against the deleterious effects of type 3 DM on cognitive brain functions.
Collapse
Affiliation(s)
- Hader Ibrahim Sakr
- Department of Medical Physiology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed A. Amen
- Department of Medical Physiology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Laila A. Rashed
- Department of Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Akef A. Khowailed
- Department of Medical Physiology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Hazem A. Sayed
- Department of Anatomy and Embryology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Moustafa E. Motawee
- Department of Histology and Cytology, Faculty of Medicine, Alazhar University, Cairo, Egypt
| | - Hany Sakr
- Department of Pathology and Laboratory Medicine, VAMC North East Ohio Healthcare System, Louis Stokes, Cleveland, Ohio, USA
| | - Mohamed Mansour Khalifa
- Department of Medical Physiology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
27
|
Flores-Dorantes MT, Díaz-López YE, Gutiérrez-Aguilar R. Environment and Gene Association With Obesity and Their Impact on Neurodegenerative and Neurodevelopmental Diseases. Front Neurosci 2020; 14:863. [PMID: 32982666 PMCID: PMC7483585 DOI: 10.3389/fnins.2020.00863] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022] Open
Abstract
Obesity is a multifactorial disease in which environmental conditions and several genes play an important role in the development of this disease. Obesity is associated with neurodegenerative diseases (Alzheimer, Parkinson, and Huntington diseases) and with neurodevelopmental diseases (autism disorder, schizophrenia, and fragile X syndrome). Some of the environmental conditions that lead to obesity are physical activity, alcohol consumption, socioeconomic status, parent feeding behavior, and diet. Interestingly, some of these environmental conditions are shared with neurodegenerative and neurodevelopmental diseases. Obesity impairs neurodevelopment abilities as memory and fine-motor skills. Moreover, maternal obesity affects the cognitive function and mental health of the offspring. The common biological mechanisms involved in obesity and neurodegenerative/neurodevelopmental diseases are insulin resistance, pro-inflammatory cytokines, and oxidative damage, among others, leading to impaired brain development or cell death. Obesogenic environmental conditions are not the only factors that influence neurodegenerative and neurodevelopmental diseases. In fact, several genes implicated in the leptin-melanocortin pathway (LEP, LEPR, POMC, BDNF, MC4R, PCSK1, SIM1, BDNF, TrkB, etc.) are associated with obesity and neurodegenerative and neurodevelopmental diseases. Moreover, in the last decades, the discovery of new genes associated with obesity (FTO, NRXN3, NPC1, NEGR1, MTCH2, GNPDA2, among others) and with neurodegenerative or neurodevelopmental diseases (APOE, CD38, SIRT1, TNFα, PAI-1, TREM2, SYT4, FMR1, TET3, among others) had opened new pathways to comprehend the common mechanisms involved in these diseases. In conclusion, the obesogenic environmental conditions, the genes, and the interaction gene-environment would lead to a better understanding of the etiology of these diseases.
Collapse
Affiliation(s)
- María Teresa Flores-Dorantes
- Laboratorio de Biología Molecular y Farmacogenómica, Centro de Investigación de Ciencia y Tecnología Aplicada de Tabasco, División Académica de Ciencias Básicas, Universidad Juárez Autónoma de Tabasco, Villahermosa, Mexico
| | - Yael Efren Díaz-López
- Laboratorio de Enfermedades Metabólicas: Obesidad y Diabetes, Hospital Infantil de México “Federico Gómez,”Mexico City, Mexico
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Ruth Gutiérrez-Aguilar
- Laboratorio de Enfermedades Metabólicas: Obesidad y Diabetes, Hospital Infantil de México “Federico Gómez,”Mexico City, Mexico
- División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| |
Collapse
|
28
|
Bolo NR, Jacobson AM, Musen G, Keshavan MS, Simonson DC. Acute Hyperglycemia Increases Brain Pregenual Anterior Cingulate Cortex Glutamate Concentrations in Type 1 Diabetes. Diabetes 2020; 69:1528-1539. [PMID: 32295804 PMCID: PMC7306132 DOI: 10.2337/db19-0936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 04/09/2020] [Indexed: 12/15/2022]
Abstract
The brain mechanisms underlying the association of hyperglycemia with depressive symptoms are unknown. We hypothesized that disrupted glutamate metabolism in pregenual anterior cingulate cortex (ACC) in type 1 diabetes (T1D) without depression affects emotional processing. Using proton MRS, we measured glutamate concentrations in ACC and occipital lobe cortex (OCC) in 13 subjects with T1D without major depression (HbA1c 7.1 ± 0.7% [54 ± 7 mmol/mol]) and 11 healthy control subjects without diabetes (HbA1c 5.5 ± 0.2% [37 ± 3 mmol/mol]) during fasting euglycemia followed by a 60-min +5.5 mmol/L hyperglycemic clamp (HG). Intrinsic neuronal activity was assessed using resting-state blood oxygen level-dependent functional MRI to measure the fractional amplitude of low-frequency fluctuations in slow-4 band (fALFF4). Emotional processing and depressive symptoms were assessed using emotional tasks (emotional Stroop task, self-referent encoding task [SRET]) and clinical ratings (Hamilton Depression Rating Scale [HAM-D], Symptom Checklist-90 Revised [SCL-90-R]), respectively. During HG, ACC glutamate increased (1.2 mmol/kg, 10% P = 0.014) while ACC fALFF4 was unchanged (-0.007, -2%, P = 0.449) in the T1D group; in contrast, glutamate was unchanged (-0.2 mmol/kg, -2%, P = 0.578) while fALFF4 decreased (-0.05, -13%, P = 0.002) in the control group. OCC glutamate and fALFF4 were unchanged in both groups. T1D had longer SRET negative word response times (P = 0.017) and higher depression rating scores (HAM-D P = 0.020, SCL-90-R depression P = 0.008). Higher glutamate change tended to associate with longer emotional Stroop response times in T1D only. Brain glutamate must be tightly controlled during hyperglycemia because of the risk for neurotoxicity with excessive levels. Results suggest that ACC glutamate control mechanisms are disrupted in T1D, which affects glutamatergic neurotransmission related to emotional or cognitive processing. Increased prefrontal glutamate during acute hyperglycemic episodes could explain our previous findings of associations among chronic hyperglycemia, cortical thinning, and depressive symptoms in T1D.
Collapse
Affiliation(s)
- Nicolas R Bolo
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA
- Department of Psychiatry, Harvard Medical School, Boston, MA
| | - Alan M Jacobson
- Research Institute, NYU Long Island School of Medicine, Mineola, NY
| | - Gail Musen
- Department of Psychiatry, Harvard Medical School, Boston, MA
- Research Division, Joslin Diabetes Center, Boston, MA
| | - Matcheri S Keshavan
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA
- Department of Psychiatry, Harvard Medical School, Boston, MA
| | - Donald C Simonson
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
29
|
Kohn JN, Troyer E, Guay-Ross RN, Wilson K, Walker A, Spoon C, Pruitt C, Lyasch G, Pung MA, Milic M, Redwine LS, Hong S. Self-reported sleep disturbances are associated with poorer cognitive performance in older adults with hypertension: a multi-parameter risk factor investigation. Int Psychogeriatr 2020; 32:815-825. [PMID: 31647051 PMCID: PMC8011648 DOI: 10.1017/s1041610219001492] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVES Given the evidence of multi-parameter risk factors in shaping cognitive outcomes in aging, including sleep, inflammation, cardiometabolism, and mood disorders, multidimensional investigations of their impact on cognition are warranted. We sought to determine the extent to which self-reported sleep disturbances, metabolic syndrome (MetS) factors, cellular inflammation, depressive symptomatology, and diminished physical mobility were associated with cognitive impairment and poorer cognitive performance. DESIGN This is a cross-sectional study. SETTING Participants with elevated, well-controlled blood pressure were recruited from the local community for a Tai Chi and healthy-aging intervention study. PARTICIPANTS One hundred forty-five older adults (72.7 ± 7.9 years old; 66% female), 54 (37%) with evidence of cognitive impairment (CI) based on Montreal Cognitive Assessment (MoCA) score ≤24, underwent medical, psychological, and mood assessments. MEASUREMENTS CI and cognitive domain performance were assessed using the MoCA. Univariate correlations were computed to determine relationships between risk factors and cognitive outcomes. Bootstrapped logistic regression was used to determine significant predictors of CI risk and linear regression to explore cognitive domains affected by risk factors. RESULTS The CI group were slower on the mobility task, satisfied more MetS criteria, and reported poorer sleep than normocognitive individuals (all p < 0.05). Multivariate logistic regression indicated that sleep disturbances, but no other risk factors, predicted increased risk of evidence of CI (OR = 2.00, 95% CI: 1.26-4.87, 99% CI: 1.08-7.48). Further examination of MoCA cognitive subdomains revealed that sleep disturbances predicted poorer executive function (β = -0.26, 95% CI: -0.51 to -0.06, 99% CI: -0.61 to -0.02), with lesser effects on visuospatial performance (β = -0.20, 95% CI: -0.35 to -0.02, 99% CI: -0.39 to 0.03), and memory (β = -0.29, 95% CI: -0.66 to -0.01, 99% CI: -0.76 to 0.08). CONCLUSIONS Our results indicate that the deleterious impact of self-reported sleep disturbances on cognitive performance was prominent over other risk factors and illustrate the importance of clinician evaluation of sleep in patients with or at risk of diminished cognitive performance. Future, longitudinal studies implementing a comprehensive neuropsychological battery and objective sleep measurement are warranted to further explore these associations.
Collapse
Affiliation(s)
- Jordan N. Kohn
- Department of Psychiatry, University of California, San Diego, USA
| | - Emily Troyer
- Department of Psychiatry, University of California, San Diego, USA
| | | | - Kathleen Wilson
- Department of Family Medicine and Public Health, University of California, San Diego, USA
| | - Amanda Walker
- Department of Family Medicine and Public Health, University of California, San Diego, USA
| | - Chad Spoon
- Department of Family Medicine and Public Health, University of California, San Diego, USA
| | - Christopher Pruitt
- Department of Family Medicine and Public Health, University of California, San Diego, USA
| | - Gary Lyasch
- Department of Psychiatry, University of California, San Diego, USA
| | - Meredith A. Pung
- Department of Family Medicine and Public Health, University of California, San Diego, USA
| | - Milos Milic
- Department of Medicine, University of California, San Diego, USA
| | | | - Suzi Hong
- Department of Psychiatry, University of California, San Diego, USA
- Department of Family Medicine and Public Health, University of California, San Diego, USA
| |
Collapse
|
30
|
Mollon J, Curran JE, Mathias SR, Knowles EEM, Carlisle P, Fox PT, Olvera RL, Göring HHH, Rodrigue A, Almasy L, Duggirala R, Blangero J, Glahn DC. Neurocognitive impairment in type 2 diabetes: evidence for shared genetic aetiology. Diabetologia 2020; 63:977-986. [PMID: 32016567 PMCID: PMC7150650 DOI: 10.1007/s00125-020-05101-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/15/2020] [Indexed: 01/02/2023]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes is associated with cognitive impairments, but it is unclear whether common genetic factors influence both type 2 diabetes risk and cognition. METHODS Using data from 1892 Mexican-American individuals from extended pedigrees, including 402 with type 2 diabetes, we examined possible pleiotropy between type 2 diabetes and cognitive functioning, as measured by a comprehensive neuropsychological test battery. RESULTS Negative phenotypic correlations (ρp) were observed between type 2 diabetes and measures of attention (Continuous Performance Test [CPT d']: ρp = -0.143, p = 0.001), verbal memory (California Verbal Learning Test [CVLT] recall: ρp = -0.111, p = 0.004) and face memory (Penn Face Memory Test [PFMT]: ρp = -0.127, p = 0.002; PFMT Delayed: ρp = -0.148, p = 2 × 10-4), replicating findings of cognitive impairment in type 2 diabetes. Negative genetic correlations (ρg) were also observed between type 2 diabetes and measures of attention (CPT d': ρg = -0.401, p = 0.001), working memory (digit span backward test: ρg = -0.380, p = 0.005), and face memory (PFMT: ρg = -0.476, p = 2 × 10-4; PFMT Delayed: ρg = -0.376, p = 0.005), suggesting that the same genetic factors underlying risk for type 2 diabetes also influence poor cognitive performance in these domains. Performance in these domains was also associated with type 2 diabetes risk using an endophenotype ranking value approach. Specifically, on measures of attention (CPT d': β = -0.219, p = 0.005), working memory (digit span backward: β = -0.326, p = 0.035), and face memory (PFMT: β = -0.171, p = 0.023; PFMT Delayed: β = -0.215, p = 0.005), individuals with type 2 diabetes showed the lowest performance, while unaffected/unrelated individuals showed the highest performance, and those related to an individual with type 2 diabetes performed at an intermediate level. CONCLUSIONS/INTERPRETATION These findings suggest that cognitive impairment may be a useful endophenotype of type 2 diabetes and, therefore, help to elucidate the pathophysiological underpinnings of this chronic disease. DATA AVAILABILITY The data analysed in this study is available in dbGaP: www.ncbi.nlm.nih.gov/projects/gap/cgi-bin/study.cgi?study_id=phs001215.v2.p2.
Collapse
Affiliation(s)
- Josephine Mollon
- Department of Psychiatry, Boston Children's Hospital, Harvard Medical School, 1 Autumn Street, BCH 3428, Boston, MA, 02115, USA.
| | - Joanne E Curran
- Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, USA
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Samuel R Mathias
- Department of Psychiatry, Boston Children's Hospital, Harvard Medical School, 1 Autumn Street, BCH 3428, Boston, MA, 02115, USA
| | - Emma E M Knowles
- Department of Psychiatry, Boston Children's Hospital, Harvard Medical School, 1 Autumn Street, BCH 3428, Boston, MA, 02115, USA
| | - Phoebe Carlisle
- Olin Neuropsychiatry Research Center, Institute of Living, Hartford, CT, USA
| | - Peter T Fox
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Rene L Olvera
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Psychiatry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Harald H H Göring
- Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, USA
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Amanda Rodrigue
- Department of Psychiatry, Boston Children's Hospital, Harvard Medical School, 1 Autumn Street, BCH 3428, Boston, MA, 02115, USA
| | - Laura Almasy
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ravi Duggirala
- Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, USA
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - John Blangero
- Department of Human Genetics, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, USA
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - David C Glahn
- Department of Psychiatry, Boston Children's Hospital, Harvard Medical School, 1 Autumn Street, BCH 3428, Boston, MA, 02115, USA
- Olin Neuropsychiatry Research Center, Institute of Living, Hartford, CT, USA
| |
Collapse
|
31
|
Association between diabetes and cognitive function at baseline in the Brazilian Longitudinal Study of Adult Health (ELSA- Brasil). Sci Rep 2020; 10:1596. [PMID: 32005901 PMCID: PMC6994611 DOI: 10.1038/s41598-020-58332-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 01/06/2020] [Indexed: 01/21/2023] Open
Abstract
Diabetes has been associated with cognitive changes and an increased risk of vascular dementia and Alzheimer’s disease, but it is unclear whether there are associations between diabetes and early alterations in cognitive performance. The present study consisted of a cross-section analysis of 14,444 participants aged 35–74 years and from a developing country at baseline in the Brazilian Longitudinal Study of Adult Health (ELSA–Brasil); these participants were recruited between 2008 and 2010. We investigated whether there was an association between diabetes and early changes in the cognitive performance of this Brazilian population. To assess cognitive domains, we used the word-list learning, word-list delayed recall and word recognition tests along. Phonemic verbal fluency tests included semantic phonemic test (animals) and a phonemic test (words beginning with the letter F). Executive functions associated with attention, concentration and psychomotor speed were evaluated using the Trail Making Test B. The exposure variable in the study was defined as diabetes. Multiple linear regression was used to estimate the association between diabetes and cognitive performance. The results were adjusted for age, sex, education, hypertension, coronary disease, depression, physical activity, smoking, alcohol consumption, and the cholesterol/HDL-C ratio. We found a significant association between diabetes and decreased memory, language and executive function (attention, concentration and psychomotor speed) performance in this population from a country with a distinct epidemiological profile, even after adjusting for the main intervening variables.
Collapse
|
32
|
Bloch K, Hornfeld SH, Dar S, Vanichkin A, Gil-Ad I, Vardi P, Weizman A. Long-term effects of intracranial islet grafting on cognitive functioning in a rat metabolic model of sporadic Alzheimer's disease-like dementia. PLoS One 2020; 15:e0227879. [PMID: 31929603 PMCID: PMC6957181 DOI: 10.1371/journal.pone.0227879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/31/2019] [Indexed: 11/24/2022] Open
Abstract
Accumulating evidence suggests that Alzheimer’s disease is associated with brain insulin resistance, as are some other types of dementia. Intranasal insulin administration has been suggested as a potential approach to overcoming brain insulin resistance and improving cognitive functions. Islet transplantation into the cranial subarachnoid cavity was used as an alternative route for insulin delivery into the brain. Recently, the authors showed the short-term beneficial cognitive effect of a small number of intracranially grafted islets in rats with cognitive dysfunction induced by intracerebroventricular administration of streptozotocin (icv-STZ). This was associated with continuous and safe insulin delivery to the rat brain. The current study investigated the long-term effect of intracranial grafting of islets on cognitive functioning in icv-STZ rats. Severe dementia, associated with obesity and cerebral amyloid-β angiopathy, was induced in Lewis inbred rats by icv-STZ. Two months after icv-STZ, one hundred syngeneic islets were transplanted into the cranial subarachnoid space. Two and six months later, cognitive alterations were assessed by Morris water-maze tests. Islet graft survival was evaluated by immunohistochemical and biochemical assays. Improvement was found in spatial learning and memory of grafted rats as opposed to the sham-operated icv-STZ rats. The grafted islets showed intact morphology, intensive expression of insulin, glucagon and glucose transporter 2. Normoglycemic obesity and cerebral amyloid-β angiopathy were found in both grafted and sham-operated icv-STZ rats. In conclusion, islet grafting into cranial subarachnoid space provides an efficient and safe approach for insulin delivery to the brain, leading to a long-term attenuation of icv-STZ-induced cognitive dysfunction.
Collapse
Affiliation(s)
- Konstantin Bloch
- Laboratory of Diabetes and Obesity Research, Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel Aviv University, Petah Tikva, Israel
- * E-mail:
| | - Shay Henry Hornfeld
- Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel Aviv University, Petah Tikva, Israel
| | - Shira Dar
- Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel Aviv University, Petah Tikva, Israel
| | - Alexey Vanichkin
- Laboratory of Transplantation, Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel Aviv University, Petah Tikva, Israel
| | - Irit Gil-Ad
- Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel Aviv University, Petah Tikva, Israel
| | - Pnina Vardi
- Laboratory of Diabetes and Obesity Research, Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel Aviv University, Petah Tikva, Israel
| | - Abraham Weizman
- Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel Aviv University, Petah Tikva, Israel
- Research Unit, Geha Mental Health Center, Petah Tikva, Israel
| |
Collapse
|
33
|
Hippocampal electrophysiological responses and changes in oxidative stress marker and serum lipid profile to pharmacological and non-pharmacological treatments of high-fat-fructose diet induced metabolic syndrome. EUROPEAN PHARMACEUTICAL JOURNAL 2019. [DOI: 10.2478/afpuc-2019-0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract
The aim of our study was to evaluate the possibility of influencing the risk factors of metabolic syndrome (MetS) and metabolic cognitive syndrome. As a model of MetS, we used high-fat-fructose diet (HFFD) fed hypertriacylglycerolemic (HTG) rats. Control group included HTG rats fed with HFFD during 8 weeks (HFFD8). Furthermore, we tested the effect of pharmacological and non-pharmacological therapies. Non-pharmacological therapy, which we chose, was a change in diet from HFFD (5 weeks) to standard one (3 weeks) and thus caloric restriction (HFFD5+3). The drug we used was rosmarinic acid (RA; 100mg/kg), which we administered to rats after 5 weeks of HFFD once a day for consecutive 3 weeks with current change in diet to standard one (HFFD5+3+RA) or during lasting last 3 weeks of HFFD (HFFD8+RA). After 8 weeks of experiment, lipid peroxidation markers, lipid profile of blood serum, and neuronal transmission and synaptic plasticity (long-term potentiation [LTP]) in hippocampal sections were evaluated in vitro. We observed a significant effect of dietary change in lipid profile (decreased total cholesterol and low-density lipoprotein cholesterol [LDL-cholesterol] and increased high-density lipoprotein cholesterol [HDL-cholesterol]). The combination of pharmacological and non-pharmacological treatments caused a decrease in total cholesterol, LDL-cholesterol, and lipid peroxidation in blood serum. Change in HFFD to standard diet without treatment resulted in slight improvement in neuronal transmission in the hippocampus and caloric restriction alone also had positive effect on LTP maintenance. Our results suggest that combination of pharmacological and non-pharmacological approaches had better impact on the biochemical parameters of MetS in blood serum, but weak impact on neuronal functions in the hippocampus, where the expected positive effect was achieved only by caloric restriction.
Collapse
|
34
|
Hovens IB, Dalenberg JR, Small DM. A Brief Neuropsychological Battery for Measuring Cognitive Functions Associated with Obesity. Obesity (Silver Spring) 2019; 27:1988-1996. [PMID: 31654505 PMCID: PMC6868337 DOI: 10.1002/oby.22644] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 08/12/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Although ample evidence links obesity to cognitive dysfunction, the trajectory of cognitive change, the underlying mechanisms, and the involvement of related factors, such as metabolic disease and diet, remain unclear. To support further investigations of BMI and cognition, this study aimed to create a concise test battery to be used in future trials. METHODS Twenty neurocognitive measures were regressed on BMI in the Human Connectome Project Healthy Young Adult S1200 data release by using linear mixed models and by adjusting for major confounders. Measures were then identified by using least absolute shrinkage and selection operator regression analysis to select tests most strongly associated with BMI. To guide further test selection, the explained variance for each variable was visualized in the final model. RESULTS BMI was negatively associated with seven neurocognitive measures. Variable selection yielded a model that included years of education and, in order of model weight, delay discounting, the relational task, the Penn Progressive Matrices test, the oral reading recognition test, the Variable Short Penn Line Orientation test, and the Penn Word Memory test. CONCLUSIONS This research resulted in an approximate 40-minute test battery for the BMI-cognition relationship in young adults that can be used in trials investigating the interrelationship between obesity and cognitive performance.
Collapse
Affiliation(s)
- Iris B. Hovens
- Modern Diet and Physiology Research Center, New Haven, CT, United States
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Jelle R. Dalenberg
- Modern Diet and Physiology Research Center, New Haven, CT, United States
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Dana M. Small
- Modern Diet and Physiology Research Center, New Haven, CT, United States
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- Department of Psychology, Yale University, New Haven, CT, United States
| |
Collapse
|
35
|
The Novel Perspectives of Adipokines on Brain Health. Int J Mol Sci 2019; 20:ijms20225638. [PMID: 31718027 PMCID: PMC6887733 DOI: 10.3390/ijms20225638] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022] Open
Abstract
First seen as a fat-storage tissue, the adipose tissue is considered as a critical player in the endocrine system. Precisely, adipose tissue can produce an array of bioactive factors, including cytokines, lipids, and extracellular vesicles, which target various systemic organ systems to regulate metabolism, homeostasis, and immune response. The global effects of adipokines on metabolic events are well defined, but their impacts on brain function and pathology remain poorly defined. Receptors of adipokines are widely expressed in the brain. Mounting evidence has shown that leptin and adiponectin can cross the blood–brain barrier, while evidence for newly identified adipokines is limited. Significantly, adipocyte secretion is liable to nutritional and metabolic states, where defective circuitry, impaired neuroplasticity, and elevated neuroinflammation are symptomatic. Essentially, neurotrophic and anti-inflammatory properties of adipokines underlie their neuroprotective roles in neurodegenerative diseases. Besides, adipocyte-secreted lipids in the bloodstream can act endocrine on the distant organs. In this article, we have reviewed five adipokines (leptin, adiponectin, chemerin, apelin, visfatin) and two lipokines (palmitoleic acid and lysophosphatidic acid) on their roles involving in eating behavior, neurotrophic and neuroprotective factors in the brain. Understanding and regulating these adipokines can lead to novel therapeutic strategies to counteract metabolic associated eating disorders and neurodegenerative diseases, thus promote brain health.
Collapse
|
36
|
Yermakov LM, Griggs RB, Drouet DE, Sugimoto C, Williams MT, Vorhees CV, Susuki K. Impairment of cognitive flexibility in type 2 diabetic db/db mice. Behav Brain Res 2019; 371:111978. [PMID: 31141724 PMCID: PMC6579681 DOI: 10.1016/j.bbr.2019.111978] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 01/05/2023]
Abstract
Impaired executive function is a major peril for patients with type 2 diabetes, reducing quality of life and ability for diabetes management. Despite the significance of this impairment, few animal models of type 2 diabetes examine domains of executive function such as cognitive flexibility or working memory. Here, we evaluated these executive function domains in db/db mice, an established model of type 2 diabetes, at 10 and 24 weeks of age. The db/db mice showed impaired cognitive flexibility in the Morris water maze reversal phase. However, the db/db mice did not show apparent working memory disturbance in the spatial working memory version of the Morris water maze or in the radial water maze. We also examined axon initial segments (AIS) and nodes of Ranvier, key axonal domains for action potential initiation and propagation. AIS were significantly shortened in medial prefrontal cortex and hippocampus of 26-week-old db/db mice compared with controls, similar to our previous findings in 10-week-old mice. Nodes of Ranvier in corpus callosum, previously shown to be unchanged at 10 weeks, were elongated at 26 weeks, suggesting an important role for this domain in disease progression. Together, the findings help establish db/db mice as a model of impaired cognitive flexibility in type 2 diabetes and advance our understanding of its pathophysiology.
Collapse
Affiliation(s)
- Leonid M Yermakov
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Ryan B Griggs
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Domenica E Drouet
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Chiho Sugimoto
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, USA
| | - Michael T Williams
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, USA
| | - Charles V Vorhees
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, USA
| | - Keiichiro Susuki
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA.
| |
Collapse
|
37
|
Carmichael O, Stuchlik P, Pillai S, Biessels GJ, Dhullipudi R, Madden-Rusnak A, Martin S, Hsia DS, Fonseca V, Bazzano L. High-Normal Adolescent Fasting Plasma Glucose Is Associated With Poorer Midlife Brain Health: Bogalusa Heart Study. J Clin Endocrinol Metab 2019; 104:4492-4500. [PMID: 31058974 PMCID: PMC6736207 DOI: 10.1210/jc.2018-02750] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/30/2019] [Indexed: 12/24/2022]
Abstract
CONTEXT It is unclear how adolescent glycemic status relates to brain health in adulthood. OBJECTIVE To assess the association between adolescent fasting plasma glucose (FPG) and MRI-based brain measures in midlife. DESIGN Between 1973 and 1992, the Bogalusa Heart Study (BHS) collected FPG from children, 3 to 18 years old, and followed up between 1992 and 2018. Cognitive tests and brain MRI were collected in 2013 to 2016 and 2018. SETTING Observational longitudinal cohort study. PARTICIPANTS Of 1298 contacted BHS participants, 74 completed screening, and 50 completed MRI. MAIN OUTCOME MEASURES Mean FPG per participant at ages <20, 20 to 40, and over 40 years old; brain white matter hyperintensity (WMH) volume, gray matter volume, and functional MRI (fMRI) activation to a Stroop task; tests of logical and working memory, executive function, and semantic fluency. RESULTS At MRI, participants were middle aged (51.3 ± 4.4 years) and predominantly female (74%) and white (74%). Mean FPG was impaired for zero, two, and nine participants in pre-20, 20 to 40, and over-40 periods. The pre-20 mean FPG above the pre-20 median value (i.e., above 83.5 mg/dL) was associated with greater WMH volume [mean difference: 0.029% of total cranial volume, CI: (0.0059, 0.052), P = 0.015] and less fMRI activation [-1.41 units (-2.78, -0.05), P = 0.043] on midlife MRI compared with below-median mean FPG. In controlling for over-40 mean FPG status did not substantially modify the associations. Cognitive scores did not differ by pre-20 mean FPG. CONCLUSIONS High-normal adolescent FPG may be associated with preclinical brain changes in midlife.
Collapse
Affiliation(s)
- Owen Carmichael
- Pennington Biomedical Research Center, Baton Rouge, Louisiana
- Correspondence and Reprint Requests: Owen Carmichael, PhD, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, Louisiana 70808. E-mail:
| | | | | | - Geert-Jan Biessels
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, University Utrecht, Utrecht, Netherlands
| | - Ram Dhullipudi
- Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | | | - Shane Martin
- Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Daniel S Hsia
- Pennington Biomedical Research Center, Baton Rouge, Louisiana
| | - Vivian Fonseca
- Section of Endocrinology, Tulane University Health Sciences Center, New Orleans, Louisiana
| | - Lydia Bazzano
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
| |
Collapse
|
38
|
Kotkowski E, Price LR, Franklin C, Salazar M, Woolsey M, DeFronzo RA, Blangero J, Glahn DC, Fox PT. A neural signature of metabolic syndrome. Hum Brain Mapp 2019; 40:3575-3588. [PMID: 31062906 PMCID: PMC6865471 DOI: 10.1002/hbm.24617] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 04/18/2019] [Accepted: 04/24/2019] [Indexed: 12/26/2022] Open
Abstract
That metabolic syndrome (MetS) is associated with age-related cognitive decline is well established. The neurobiological changes underlying these cognitive deficits, however, are not well understood. The goal of this study was to determine whether MetS is associated with regional differences in gray-matter volume (GMV) using a cross-sectional, between-group contrast design in a large, ethnically homogenous sample. T1-weighted MRIs were sampled from the genetics of brain structure (GOBS) data archive for 208 Mexican-American participants: 104 participants met or exceeded standard criteria for MetS and 104 participants were age- and sex-matched metabolically healthy controls. Participants ranged in age from 18 to 74 years (37.3 ± 13.2 years, 56.7% female). Images were analyzed in a whole-brain, voxel-wise manner using voxel-based morphometry (VBM). Three contrast analyses were performed, a whole sample analysis of all 208 participants, and two post hoc half-sample analyses split by age along the median (35.5 years). Significant associations between MetS and decreased GMV were observed in multiple, spatially discrete brain regions including the posterior cerebellum, brainstem, orbitofrontal cortex, bilateral caudate nuclei, right parahippocampus, right amygdala, right insula, lingual gyrus, and right superior temporal gyrus. Age, as shown in the post hoc analyses, was demonstrated to be a significant covariate. A further functional interpretation of the structures exhibiting lower GMV in MetS reflected a significant involvement in reward perception, emotional valence, and reasoning. Additional studies are needed to characterize the influence of MetS's individual clinical components on brain structure and to explore the bidirectional association between GMV and MetS.
Collapse
Affiliation(s)
- Eithan Kotkowski
- Research Imaging Institute, University of Texas Health Science Center at San AntonioSan AntonioTexas
- Department of RadiologyUniversity of Texas Health Science Center at San AntonioSan AntonioTX
| | - Larry R. Price
- Methodology, Measurement and Statistical Analysis CenterTexas State UniversitySan MarcosTexas
| | - Crystal Franklin
- Research Imaging Institute, University of Texas Health Science Center at San AntonioSan AntonioTexas
| | - Maximino Salazar
- Research Imaging Institute, University of Texas Health Science Center at San AntonioSan AntonioTexas
| | - Mary Woolsey
- Research Imaging Institute, University of Texas Health Science Center at San AntonioSan AntonioTexas
| | - Ralph A. DeFronzo
- Texas Diabetes InstituteSan AntonioTexas
- Diabetes Research Unit and Diabetes DivisionUniversity of Texas Health Science Center at San AntonioSan AntonioTexas
| | - John Blangero
- Genomics Computing Center, South Texas Diabetes and Obesity InstituteUniversity of Texas Rio Grande ValleyBrownsvilleTexas
| | - David C. Glahn
- Department of PsychiatryYale University School of MedicineNew HavenConnecticut
- Olin Neuropsychiatry Research CenterInstitute of Living, Hartford HospitalHartfordConnecticut
| | - Peter T. Fox
- Research Imaging Institute, University of Texas Health Science Center at San AntonioSan AntonioTexas
- Department of RadiologyUniversity of Texas Health Science Center at San AntonioSan AntonioTX
| |
Collapse
|
39
|
Doucet GE, Rasgon N, McEwen BS, Micali N, Frangou S. Elevated Body Mass Index is Associated with Increased Integration and Reduced Cohesion of Sensory-Driven and Internally Guided Resting-State Functional Brain Networks. Cereb Cortex 2019; 28:988-997. [PMID: 28119342 DOI: 10.1093/cercor/bhx008] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Indexed: 12/11/2022] Open
Abstract
Elevated body mass index (BMI) is associated with increased multi-morbidity and mortality. The investigation of the relationship between BMI and brain organization has the potential to provide new insights relevant to clinical and policy strategies for weight control. Here, we quantified the association between increasing BMI and the functional organization of resting-state brain networks in a sample of 496 healthy individuals that were studied as part of the Human Connectome Project. We demonstrated that higher BMI was associated with changes in the functional connectivity of the default-mode network (DMN), central executive network (CEN), sensorimotor network (SMN), visual network (VN), and their constituent modules. In siblings discordant for obesity, we showed that person-specific factors contributing to obesity are linked to reduced cohesiveness of the sensory networks (SMN and VN). We conclude that higher BMI is associated with widespread alterations in brain networks that balance sensory-driven (SMN, VN) and internally guided (DMN, CEN) states which may augment sensory-driven behavior leading to overeating and subsequent weight gain. Our results provide a neurobiological context for understanding the association between BMI and brain functional organization while accounting for familial and person-specific influences.
Collapse
Affiliation(s)
- Gaelle E Doucet
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, USA
| | - Natalie Rasgon
- Center for Neuroscience in Women's Health, Stanford University, Palo Alto, CA 91304, USA
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA
| | - Nadia Micali
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, USA
| | - Sophia Frangou
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, USA
| |
Collapse
|
40
|
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
|
41
|
Unexpected systemic phenotypes result from focal combined deficiencies of forebrain insulin receptor/IGF-1 receptor signaling. Proc Natl Acad Sci U S A 2019; 116:5852-5854. [PMID: 30858326 DOI: 10.1073/pnas.1901970116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
42
|
Luquet SH, Vaudry H, Granata R. Editorial: Neuroendocrine Control of Feeding Behavior. Front Endocrinol (Lausanne) 2019; 10:399. [PMID: 31297088 PMCID: PMC6593060 DOI: 10.3389/fendo.2019.00399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/05/2019] [Indexed: 11/13/2022] Open
Affiliation(s)
- Serge H. Luquet
- Paris Diderot University, Paris, France
- *Correspondence: Serge H. Luquet
| | - Hubert Vaudry
- Université de Rouen, Mont-Saint-Aignan, France
- Hubert Vaudry
| | - Riccarda Granata
- Department of Medical Sciences, University of Turin, Turin, Italy
- Riccarda Granata
| |
Collapse
|
43
|
Parahippocampal gyrus expression of endothelial and insulin receptor signaling pathway genes is modulated by Alzheimer's disease and normalized by treatment with anti-diabetic agents. PLoS One 2018; 13:e0206547. [PMID: 30383799 PMCID: PMC6211704 DOI: 10.1371/journal.pone.0206547] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 10/15/2018] [Indexed: 12/14/2022] Open
Abstract
A large body of literature links risk of cognitive decline, mild cognitive impairment (MCI) and dementia with Type 2 Diabetes (T2D) or pre-diabetes. Accumulating evidence implicates a close relationship between the brain insulin receptor signaling pathway (IRSP) and the accumulation of amyloid beta and hyperphosphorylated and conformationally abnormal tau. We showed previously that the neuropathological features of Alzheimer's disease (AD were reduced in patients with diabetes who were treated with insulin and oral antidiabetic medications. To understand better the neurobiological substrates of T2D and T2D medications in AD, we examined IRSP and endothelial cell markers in the parahippocampal gyrus of controls (N = 30), of persons with AD (N = 19), and of persons with AD and T2D, who, in turn, had been treated with anti-diabetic drugs (insulin and or oral agents; N = 34). We studied the gene expression of selected members of the IRSP and selective endothelial cell markers in bulk postmortem tissue from the parahippocampal gyrus and in endothelial cell enriched isolates from the same brain region. The results indicated that there are considerable abnormalities and reductions in gene expression (bulk tissue homogenates and endothelial cell isolates) in the parahippocampal gyri of persons with AD that map directly to genes associated with the microvasculature and the IRSP. Our results also showed that the numbers of abnormally expressed microvasculature and IRSP associated genes in diabetic AD donors who had been treated with anti-diabetic agents were reduced significantly. These findings suggest that anti-diabetic treatments may reduce or normalize compromised microvascular and IRSP functions in AD.
Collapse
|
44
|
Moslehi A, Hamidi-zad Z. Role of SREBPs in Liver Diseases: A Mini-review. J Clin Transl Hepatol 2018; 6:332-338. [PMID: 30271747 PMCID: PMC6160306 DOI: 10.14218/jcth.2017.00061] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 01/10/2018] [Accepted: 02/11/2018] [Indexed: 12/15/2022] Open
Abstract
Sterol regulator element binding proteins (SREBPs) are a family of transcription factors involved in the biogenesis of cholesterol, fatty acids and triglycerides. They also regulate physiological functions of many organs, such as thyroid, brain, heart, pancreas and hormone synthesis. Beside the physiological effects, SREBPs participate in some pathological processes, diabetes, endoplasmic reticulum stress, atherosclerosis and chronic kidney disease associated with SREBP expression changes. In the liver, SREBPs are involved in the pathogenesis of nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, hepatitis and hepatic cancer. There are several SREBP inhibitors that have potential for treating obesity, diabetes and cancer. This review assesses the recent findings about the roles of SREBPs in the physiology of organs' function and pathogenesis of liver diseases.
Collapse
Affiliation(s)
- Azam Moslehi
- Department of Physiology, Cellular & Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Zeinab Hamidi-zad
- Department of Physiology, Cellular & Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| |
Collapse
|
45
|
Silveira PP, Pokhvisneva I, Gaudreau H, Rifkin-Graboi A, Broekman BFP, Steiner M, Levitan R, Parent C, Diorio J, Meaney MJ. Birth weight and catch up growth are associated with childhood impulsivity in two independent cohorts. Sci Rep 2018; 8:13705. [PMID: 30209275 PMCID: PMC6135839 DOI: 10.1038/s41598-018-31816-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 08/28/2018] [Indexed: 02/07/2023] Open
Abstract
Individuals born after intrauterine growth restriction (IUGR) are more impulsive towards palatable foods, but it is not clear 1) if IUGR-related impulsivity is specific for foods and solely based on response inhibition and 2) if the development of impulsivity is due to being born IUGR per se or to growing up fast in the first few years of life (catch up growth). Children were classified in the IUGR group if the birth weight ratio was below 0.85. Delta z score for BMI was used as a measure of catch up growth. In MAVAN (N = 274), impulsivity was measured by the Information Sampling Task from the Cambridge Neuropsychological Test Automated Battery (IST - CANTAB), and in GUSTO using the Sticker Delay Task (N = 327). There is a significant effect of interaction between being born IUGR and the magnitude of catch up growth on the reflection impulsivity from IST-CANTAB at 60 months, in which greater catch up growth associates with greater impulsivity in the IST fixed condition in IUGR children. The finding was reproduced in children from the GUSTO cohort using the Sticker Delay Task. We confirmed that catch up growth interacts with IUGR, having a major role in the development of impulsivity in the first years of life and influencing inhibitory control and decision making processes.
Collapse
Affiliation(s)
- Patrícia P Silveira
- Department of Psychiatry, McGill University & Sackler Institute for Epigenetics & Psychobiology at McGill University, Montreal, Quebec, Canada.
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, H4H 1R3, Canada.
| | - Irina Pokhvisneva
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, H4H 1R3, Canada
| | - Hélène Gaudreau
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, H4H 1R3, Canada
| | - Anne Rifkin-Graboi
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Birit F P Broekman
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Meir Steiner
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, L8N 3K7, Canada
| | - Robert Levitan
- Department of Psychiatry, University of Toronto and Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8, Canada
| | - Carine Parent
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, H4H 1R3, Canada
| | - Josie Diorio
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, H4H 1R3, Canada
| | - Michael J Meaney
- Department of Psychiatry, McGill University & Sackler Institute for Epigenetics & Psychobiology at McGill University, Montreal, Quebec, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, H4H 1R3, Canada
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
46
|
Moreno CL, Della Guardia L, Shnyder V, Ortiz-Virumbrales M, Kruglikov I, Zhang B, Schadt EE, Tanzi RE, Noggle S, Buettner C, Gandy S. iPSC-derived familial Alzheimer's PSEN2 N141I cholinergic neurons exhibit mutation-dependent molecular pathology corrected by insulin signaling. Mol Neurodegener 2018; 13:33. [PMID: 29945658 PMCID: PMC6020427 DOI: 10.1186/s13024-018-0265-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 05/27/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) is a recognized risk factor for the development of cognitive impairment (CI) and/or dementia, although the exact nature of the molecular pathology of T2D-associated CI remains obscure. One link between T2D and CI might involve decreased insulin signaling in brain and/or neurons in either animal or postmortem human brains as has been reported as a feature of Alzheimer's disease (AD). Here we asked if neuronal insulin resistance is a cell autonomous phenomenon in a familial form of AD. METHODS We have applied a newly developed protocol for deriving human basal forebrain cholinergic neurons (BFCN) from skin fibroblasts via induced pluripotent stem cell (iPSC) technology. We generated wildtype and familial AD mutant PSEN2 N141I (presenilin 2) BFCNs and assessed if insulin signaling, insulin regulation of the major AD proteins Aβ and/or tau, and/or calcium fluxes is altered by the PSEN2 N141I mutation. RESULTS We report herein that wildtype, PSEN2 N141I and CRISPR/Cas9-corrected iPSC-derived BFCNs (and their precursors) show indistinguishable insulin signaling profiles as determined by the phosphorylation of canonical insulin signaling pathway molecules. Chronic insulin treatment of BFCNs of all genotypes led to a reduction in the Aβ42/40 ratio. Unexpectedly, we found a CRISPR/Cas9-correctable effect of PSEN2 N141I on calcium flux, which could be prevented by chronic exposure of BFCNs to insulin. CONCLUSIONS Our studies indicate that the familial AD mutation PSEN2 N141I does not induce neuronal insulin resistance in a cell autonomous fashion. The ability of insulin to correct calcium fluxes and to lower Aβ42/40 ratio suggests that insulin acts to oppose an AD-pathophysiology. Hence, our results are consistent with a potential physiological role for insulin as a mediator of resilience by counteracting specific metabolic and molecular features of AD.
Collapse
Affiliation(s)
- Cesar L Moreno
- Department of Neurology, NFL Neurological Care Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.,Department of Psychiatry, Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, 5 E 98th St, New York, NY, 10029, USA
| | - Lucio Della Guardia
- Department of Medicine, Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Valeria Shnyder
- Department of Medicine, Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Maitane Ortiz-Virumbrales
- Department of Neurology, NFL Neurological Care Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ilya Kruglikov
- New York Stem Cell Foundation Laboratories, New York, NY, 10032, USA
| | - Bin Zhang
- Icahn Institute and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Eric E Schadt
- Icahn Institute and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rudolph E Tanzi
- Genetics and Aging Unit, Harvard Medical School, Massachusetts General Hospital, 114 16th Street, Charlestown, MA, 02129, USA
| | - Scott Noggle
- New York Stem Cell Foundation Laboratories, New York, NY, 10032, USA.
| | - Christoph Buettner
- Department of Medicine, Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Sam Gandy
- Department of Neurology, NFL Neurological Care Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA. .,Department of Psychiatry, Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, 5 E 98th St, New York, NY, 10029, USA.
| |
Collapse
|
47
|
Bugge A, Möller S, Westfall DR, Tarp J, Gejl AK, Wedderkopp N, Hillman CH. Associations between waist circumference, metabolic risk and executive function in adolescents: A cross-sectional mediation analysis. PLoS One 2018; 13:e0199281. [PMID: 29912925 PMCID: PMC6005548 DOI: 10.1371/journal.pone.0199281] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 06/05/2018] [Indexed: 12/12/2022] Open
Abstract
The main objective of this study was to investigate the associations between waist circumference, metabolic risk factors, and executive function in adolescents. Methods: The study was cross-sectional and included 558 adolescents (mean age 14.2 years). Anthropometrics and systolic blood pressure (sysBP) were measured and fasting blood samples were analyzed for metabolic risk factors. A metabolic risk factor cluster score (MetS-cluster score) was computed from the sum of standardized sysBP, triglycerides (TG), inverse high-density lipid cholesterol (HDLc) and insulin resistance (homeostasis model assessment). Cognitive control was measured with a modified flanker task. Results: Regression analyses indicated that after controlling for demographic variables, HDLc exhibited a negative and TG a positive association with flanker reaction time (RT). Waist circumference did not demonstrate a statistically significant total association with the cognitive outcomes. In structural equation modeling, waist circumference displayed an indirect positive association with incongruent RT through a higher MetS-cluster score and through lower HDLc. The only statistically significant direct association between waist circumference and the cognitive outcomes was for incongruent RT in the model including HDLc as mediator. Conclusions: These findings are consonant with the previous literature reporting an adverse association between certain metabolic risk factors and cognitive control. Accordingly, these results suggest specificity between metabolic risk factors and cognitive control outcomes. Further, results of the present study, although cross-sectional, provide new evidence that specific metabolic risk factors may mediate an indirect association between adiposity and cognitive control in adolescents, even though a direct association between these variables was not observed. However, taking the cross-sectional study design into consideration, these results should be interpreted with caution and future longitudinal or experimental studies should verify the findings of this study.
Collapse
Affiliation(s)
- Anna Bugge
- Centre of Research in Childhood Health, Institute for Sport Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
- Department of Physiotherapy and Occupational Therapy, University College Copenhagen, Copenhagen, Denmark
- * E-mail:
| | - Sören Möller
- OPEN–Odense Patient Data Explorative Network, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Daniel R. Westfall
- Department of Psychology, Northeastern University, Boston MA, United States of America
| | - Jakob Tarp
- Centre of Research in Childhood Health, Institute for Sport Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Anne K. Gejl
- Centre of Research in Childhood Health, Institute for Sport Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Niels Wedderkopp
- Centre of Research in Childhood Health, Institute for Sport Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
- Sports Medicine clinic Orthopedic Department, Middelfart Hospital, Institute of Regional Health Research, University of Southern Denmark, Middelfart, Denmark
| | - Charles H. Hillman
- Department of Psychology, Northeastern University, Boston MA, United States of America
- Department of Physical Therapy, Movement, & Rehabilitation Sciences, Northeastern University, Boston MA, United States of America
| |
Collapse
|
48
|
Yermakov LM, Drouet DE, Griggs RB, Elased KM, Susuki K. Type 2 Diabetes Leads to Axon Initial Segment Shortening in db/db Mice. Front Cell Neurosci 2018; 12:146. [PMID: 29937715 PMCID: PMC6002488 DOI: 10.3389/fncel.2018.00146] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 05/14/2018] [Indexed: 01/09/2023] Open
Abstract
Cognitive and mood impairments are common central nervous system complications of type 2 diabetes, although the neuronal mechanism(s) remains elusive. Previous studies focused mainly on neuronal inputs such as altered synaptic plasticity. Axon initial segment (AIS) is a specialized functional domain within neurons that regulates neuronal outputs. Structural changes of AIS have been implicated as a key pathophysiological event in various psychiatric and neurological disorders. Here we evaluated the structural integrity of the AIS in brains of db/db mice, an established animal model of type 2 diabetes associated with cognitive and mood impairments. We assessed the AIS before (5 weeks of age) and after (10 weeks) the development of type 2 diabetes, and after daily exercise treatment of diabetic condition. We found that the development of type 2 diabetes is associated with significant AIS shortening in both medial prefrontal cortex and hippocampus, as evident by immunostaining of the AIS structural protein βIV spectrin. AIS shortening occurs in the absence of altered neuronal and AIS protein levels. We found no change in nodes of Ranvier, another neuronal functional domain sharing a molecular organization similar to the AIS. This is the first study to identify AIS alteration in type 2 diabetes condition. Since AIS shortening is known to lower neuronal excitability, our results may provide a new avenue for understanding and treating cognitive and mood impairments in type 2 diabetes.
Collapse
Affiliation(s)
- Leonid M Yermakov
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Domenica E Drouet
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Ryan B Griggs
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Khalid M Elased
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Keiichiro Susuki
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| |
Collapse
|
49
|
Pruzin JJ, Nelson PT, Abner EL, Arvanitakis Z. Review: Relationship of type 2 diabetes to human brain pathology. Neuropathol Appl Neurobiol 2018; 44:347-362. [PMID: 29424027 PMCID: PMC5980704 DOI: 10.1111/nan.12476] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 01/12/2018] [Indexed: 12/14/2022]
Abstract
Type 2 diabetes (T2D) and Alzheimer's disease (AD) are both highly prevalent diseases worldwide, and each is associated with high-morbidity and high-mortality. Numerous clinical studies have consistently shown that T2D confers a two-fold increased risk for a dementia, including dementia attributable to AD. Yet, the mechanisms underlying this relationship, especially nonvascular mechanisms, remain debated. Cerebral vascular disease (CVD) is likely to be playing a role. But increased AD neuropathologic changes (ADNC), specifically neuritic amyloid plaques (AP) and neurofibrillary tangles (NFT), are also posited mechanisms. The clinicopathological studies to date demonstrate T2D to be consistently associated with infarcts, particularly subcortical lacunar infarcts, but not ADNC, suggesting the association of T2D with dementia may largely be mediated through CVD. Furthermore, growing interest exists in insulin resistance (IR), particularly IR within the brain itself, which may be an associated but distinct phenomenon from T2D, and possibly itself associated with ADNC. Other mechanisms largely related to protein processing and efflux in the central nervous system with altered function in T2D may also be involved. Such mechanisms include islet amyloid polypeptide (or amylin) deposition, co-localized with beta-amyloid and found in more abundance in the AD temporal cortex, blood-brain barrier breakdown and dysfunction, potentially related to pericyte degeneration, and disturbance of brain lymphatics, both in the glial lymphatic system and the newly discovered discrete central nervous system lymph vessels. Medical research is ongoing to further disentangle the relationship of T2D to dementia in the ageing human brain.
Collapse
Affiliation(s)
- Jeremy J. Pruzin
- Rush Alzheimer’s Disease Center, Chicago, IL
- Dept of Neurological Sciences, Rush University Medical Center, Chicago, IL
| | - Peter T. Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY
- Department of Pathology, University of Kentucky, Lexington, KY
| | - Erin L. Abner
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY
- Department of Epidemiology, University of Kentucky, Lexington, KY
| | - Zoe Arvanitakis
- Rush Alzheimer’s Disease Center, Chicago, IL
- Dept of Neurological Sciences, Rush University Medical Center, Chicago, IL
| |
Collapse
|
50
|
Abstract
This article reviews current treatment strategies and recent advances for the Lewy body dementias (LBDs). Current available symptom treatment strategies are based on monoaminergic, cholinergic and glutaminergic neurotransmitter systems. Relatively robust evidence exists for cholinesterase inhibitors for cognitive impairment in LBD and in Parkinson's disease for antidepressants, clozapine and recently pimavanserin for psychosis. interpidine (RVT 101) and nelotanserin are currently under investigation. Non-pharmacological interventions, such as cognitive stimulation, physical exercises and neuromodulation strategies, may be useful in Parkinson's disease but have not yet been tested in dementias. Disease-modifying approaches are aimed at preventing, slowing or ameliorating the production, aggregation and deposition of pathological proteins, including immunotherapy targeting α-synuclein and an ongoing trial using ambroxol which increases glucocerebrosidase activity to lower the levels of the protein alpha-synuclein. Other disease-modifying clinical trials are using agents to augment insulin signalling, stem cell therapy, reducing amyloid pathology and gene therapy.
Collapse
|