1
|
Yang S, Zhang H, Zhu W, Niu T, Farooqui HFM, Wang J, Yang M, Liu E, Wang S. Peripheral blood heat shock protein 27 correlates with information processing speed and executive function, potentially serving as a marker for mild cognitive impairment in patients with type 2 diabetes mellitus. Diabetol Metab Syndr 2025; 17:167. [PMID: 40410804 DOI: 10.1186/s13098-025-01747-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/16/2025] [Indexed: 05/25/2025] Open
Abstract
BACKGROUND AND AIMS Previous study found that interleukin 1β (IL-1β) is associated with diabetic cognitive dysfunction. Heat shock protein 27 (HSP27) is one of the factors related to IL-1β associated inflammation. Here, we aim to investigate the role of HSP27 in mild cognitive impairment (MCI) in patients with type 2 diabetes mellitus (T2DM). MATERIALS AND METHODS In this study, individuals with T2DM with and without MCI were recruited and categorized into Control and MCI groups. Plasma HSP27 levels were assessed and compared between the Control and MCI groups. Furthermore, the relationship between HSP27 and diabetic dysfunction was elucidated through association and regression analyses. Finally, diagnostic values were determined using ROC curves. RESULTS In humans, individuals with T2DM and MCI exhibit decreased levels of HSP27 compared to those without MCI. Notably, the levels of HSP27 are associated with neuropsychological test scores that reflect cognitive preferences. Additionally, decreased HSP27 levels serve as one of the risk factors for MCI in T2DM patients (OR = 0.355, P = 0.002). Moreover, there is a defined cut-off point for HSP27 in diagnosing MCI, set at 3.51 pg/ml, with a sensitivity of 47.2%, a specificity of 94.4%, and an area under the curve (AUC) of 0.695. CONCLUSIONS Generally speaking, HSP27 is linked to cognitive decline in individuals with T2DM. Decreased levels of HSP27 in plasma are identified as both a risk factor for MCI and a potential diagnostic biomarker for MCI in patients with T2DM. The diagnostic value of HSP27 in MCI is primarily reflected in its demonstrated true negative rate.
Collapse
Affiliation(s)
- Shufang Yang
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
- Department of Endocrinology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Haoqiang Zhang
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Wenwen Zhu
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Tong Niu
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | | | - Jue Wang
- Department of Endocrinology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
- Medical school, Dalian Medical University, Dalian, China
| | - Mingyue Yang
- Department of Endocrinology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
- Medical school, Dalian Medical University, Dalian, China
| | - Enlin Liu
- Department of Endocrinology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
- Medical school, Nantong University, Nantong, China
| | - Shaohua Wang
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China.
| |
Collapse
|
2
|
Cohen A, Levine SZ, Vainstein G, Beeri MS, Weinstein G. New-generation antidiabetic medications and dementia risk in older adults with type 2 diabetes: A retrospective cohort study. J Prev Alzheimers Dis 2025:100199. [PMID: 40345929 DOI: 10.1016/j.tjpad.2025.100199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 04/24/2025] [Accepted: 04/30/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND New-generation antidiabetic medications may have therapeutic potential for dementia, beyond their glycemic effects. However, information from observational studies exploring the association between new-generation antidiabetic use and dementia risk is limited. OBJECTIVES To examine the association between new-generation antidiabetic medication use and dementia risk. DESIGN Retrospective cohort study using electronic health records of a large non-profit health maintenance organization. PARTICIPANTS 84,798 dementia-free individuals aged ≥65y with type 2 diabetes. MEASUREMENTS Antidiabetic medication exposure was based on purchased prescriptions and was used as a time-varying variable. Exposure periods were defined as periods in which either dipeptidyl peptidase-4 inhibitors (DPP-4i), sodium-glucose cotransporter-2 inhibitors (SGLT-2i), or glucagon-like peptide-1 analogs (GLP-1a) or their combinations were used, otherwise unexposed. Dementia classification was based on the International Classification of Diseases, Ninth Revision codes or antidementia medication prescriptions. Cox regression models were fitted to quantify the association between antidiabetic medication use and incident dementia. Models were adjusted for 13 potential sources of confounding using inverse-probability weighting. RESULTS Among 84,798 individuals with a mean diabetes onset age of 66.4 ± 7.5 years, the median follow-up for dementia risk was 8.7 years (Q1-Q3: 5.4-12.8). Dementia was diagnosed in 11,642 (13.7%) individuals. New-generation medication use was associated with reduced dementia risk (HR = 0.69; 95% CI, 0.66-0.73) and by drug classes (DPP-4i, HR 0.67 [95% CI 0.63-0.71]; SGLT-2i, 0.63 [95% CI 0.56-0.70], GLP-1a, 0.61 [95% CI 0.54-0.69]. CONCLUSIONS The results of this large-scale study suggest that new-generation antidiabetic medication use may be associated with lower dementia risk in older adults with T2D.
Collapse
Affiliation(s)
- Avi Cohen
- School of Public Health, University of Haifa, Haifa 3498838, Israel.
| | - Stephen Z Levine
- School of Public Health, University of Haifa, Haifa 3498838, Israel
| | - Gabriel Vainstein
- Kahn-Sagol-Maccabi Research and Innovation Institute, Tel-Aviv 6800001, Israel
| | - Michal Schnaider Beeri
- The Kreiger Klein Alzheimer's Research Center, Brain Health Institute, Rutgers Health, New Brunswick 08901, NJ, USA
| | - Galit Weinstein
- School of Public Health, University of Haifa, Haifa 3498838, Israel
| |
Collapse
|
3
|
Cagigas ML, De Ciutiis I, Masedunskas A, Fontana L. Dietary and pharmacological energy restriction and exercise for healthspan extension. Trends Endocrinol Metab 2025:S1043-2760(25)00076-1. [PMID: 40318928 DOI: 10.1016/j.tem.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 03/30/2025] [Accepted: 04/02/2025] [Indexed: 05/07/2025]
Abstract
Extending healthspan - the years lived in optimal health - holds transformative potential to reduce chronic diseases and healthcare costs. Dietary restriction (DR), particularly when combined with nutrient-rich diets and exercise, is among the most effective, evidence-based strategies for enhancing metabolic health and longevity. By targeting fundamental pathways, it mitigates the onset and progression of obesity, type 2 diabetes (T2D), cardiovascular disease (CVD), neurodegeneration, and cancer. This review synthesizes human data on the impact of DR and exercise on metabolic and age-related diseases, while emphasizing key biological mechanisms such as nutrient sensing, insulin sensitivity, inflammation, mitochondrial function, and gut microbiota. We also examine the emerging role of pharmacologically induced DR, focusing on glucagon-like peptide 1 (GLP-1) receptor agonists (RAs) that partially mimic DR and present opportunities for chronic disease prevention.
Collapse
Affiliation(s)
- Maria Lastra Cagigas
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Isabella De Ciutiis
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Andrius Masedunskas
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Luigi Fontana
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW, Australia.
| |
Collapse
|
4
|
Chen YY, Chen YH, Fang YW, Wang JT, Tsai MH. The protective effects of insulin on the developing of dementia in chronic kidney disease patients with hypertension and diabetes: a population-based nationwide study. BMC Nephrol 2025; 26:211. [PMID: 40281491 PMCID: PMC12032640 DOI: 10.1186/s12882-025-04145-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 04/21/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Chronic kidney disease (CKD), hypertension, and diabetes are associated with dementia, and insulin resistance promotes vascular dysfunction resulting in dementia. However, the study of insulin use in preventing dementia in CKD patients with diabetes and hypertension is limited. We aim to assess the effects of insulin use on the incidence of dementia in patients with CKD with hypertension and diabetes. DESIGN, SETTING AND PARTICIPANTS A retrospective cohort study using the nationwide database from Taiwan's National Health Insurance Research Database. We selected 11,758 CKD patients with diabetes and hypertension in 2006, including 5,864 insulin users and 5,894 non-insulin users. Moreover, their medication possession ratios (MPR) were calculated. MAIN OUTCOMES We used the competing risk model to estimate the hazard ratio (HR) for the incidence of dementia for insulin use in the target population. RESULTS In a follow-up period of 11 years, 1285 events of dementia were recorded, and the multivariate-adjusted HR for dementia by insulin usage (yes vs. no) and insulin usage per MPR is 0.652 (95% confidence interval [CI]: 0.552 to 0.771) and 0.995 (95% CI: 0.993 to 0.998) respectively. Such a significant negative association was consistent in almost all subgroups. Moreover, a dose-dependent effect of insulin was noted, where patients with higher insulin MPRs were less likely to have dementia. CONCLUSION The CKD patients with hypertension and diabetes who received insulin therapy had a 35% decreased risk of dementia.
Collapse
Affiliation(s)
- Yun-Yi Chen
- Department of Research, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- Institute of Hospital and Health Care Administration, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Hsien Chen
- Department of Family Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Yu-Wei Fang
- Division of Nephrology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- Department of Medicine, Fu-Jen Catholic University School of Medicine, Taipei, Taiwan
| | - Jing-Tong Wang
- Division of Nephrology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Ming-Hsien Tsai
- Division of Nephrology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan.
- Department of Medicine, Fu-Jen Catholic University School of Medicine, Taipei, Taiwan.
| |
Collapse
|
5
|
Hui EK, Mukadam N, Kohl G, Livingston G. Effect of diabetes medications on the risk of developing dementia, mild cognitive impairment, or cognitive decline: A systematic review and meta-analysis. J Alzheimers Dis 2025; 104:627-648. [PMID: 40017057 DOI: 10.1177/13872877251319054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Background: Diabetes is a risk factor for dementia, but we do not know whether specific diabetes medications ameliorate this risk. Objective: To systematically review and meta-analyze such medication's effect on the risk of developing dementia, mild cognitive impairment (MCI), or cognitive decline. Methods: We searched three databases until 21 November 2023. We included randomized controlled trials (RCT), cohort, and case-control studies assessing association between antidiabetic medication and future dementia, MCI, or cognitive decline. We meta-analyzed studies separately for individual drug classes and their comparators (no medication, placebo, or another drug). We appraised study quality using the Newcastle-Ottawa Scale and Physiotherapy Evidence Database Scale. Results: 42 studies fulfilled inclusion criteria. Glucagon-like peptide-1 receptor agonists (GLP-1 RA) versus placebo reduced dementia risk by 53% in three RCTs (n = 15,820, RR = 0.47[0.25, 0.86]) and 27% in three case-control studies (n = 312,856, RR = 0.73[0.54, 0.99], I2 = 96%). Repaglinide was superior to glibenclamide by 0.8 points on the Mini-Mental State Examination scale in another RCT. Meta-analysis of seven longitudinal studies showed glitazones (n = 1,081,519, RR = 0.78[0.76, 0.81], I2 = 0%) were associated with reduced dementia risk. Metformin (n = 999,349, RR = 0.94[0.79, 1.13], I2 = 98.4%), sulfonylureas (RR = 0.98[0.78, 1.22], I2 = 83.3%), dipeptidyl peptidase-IV inhibitors (DPP-1V) (n = 192,802, RR = 0.86[0.65, 1.15], I2 = 92.9%) and insulin (n = 571,274, RR = 1.09[0.95, 1.25], I2 = 94.8%) were not. Most studies were observational and limited by confounding by indication. Conclusions: In people with diabetes, RCTs consistently showed GLP-RAs reduce future dementia risk. Glitazones consistently showed protective effects, without heterogeneity, suggesting potential generalizability of these results. Metformin, sulfonylureas, insulin, and DPP-1V studies had inconsistent findings. If information is available future studies should consider dosage, severity, and duration.
Collapse
Affiliation(s)
- Esther K Hui
- Division of Psychiatry, University College London, London, UK
| | - Naaheed Mukadam
- Division of Psychiatry, University College London, London, UK
| | - Gianna Kohl
- Division of Psychology and Language Sciences, University College London, London, UK
| | - Gill Livingston
- Division of Psychiatry, University College London, London, UK
| |
Collapse
|
6
|
García-Lluch G, Marseglia A, Royo LM, Albiach JP, Garcia-Zamora M, Baquero M, Peña-Bautista C, Álvarez L, Westman E, Cháfer-Pericás C. Associations between antidiabetic medications and cerebrospinal fluid biomarkers of Alzheimer's disease. J Alzheimers Dis 2025; 103:758-774. [PMID: 39686618 DOI: 10.1177/13872877241304995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
BACKGROUND It has been hypothesized that insulin resistance is pivotal in mediating amyloid and tau dysregulations in Alzheimer's disease (AD). OBJECTIVE To investigate the impact of different antidiabetic agents, their daily dosage intake, and treatment duration on cerebrospinal fluid (CSF) AD biomarkers among patients with type 2 diabetes. METHODS This cross-sectional study selected patients between 50 and 80 years with diabetes and CSF AD biomarkers screened between 2017 and 2023 in the VALCODIS Cohort. CSF biomarkers were total tau (t-tau), phosphorylated tau 181 (p-tau), and amyloid-β 42 (Aβ42). Analytical variables were obtained. Antidiabetic prescriptions were recorded in defined daily doses (DDD), according to the ATC/DDD 2021 system, and years of drug exposure duration before lumbar puncture. Logistic regressions were performed to establish the correlations between drug usage and AD biomarker alteration. RESULTS Among patients with diabetes, Insulin consumption was associated with lower odds of abnormal Aβ42 levels (OR 0.36 [95% CI 0.15, 0.76]) and tau pathology (OR 0.49 [95% CI 0.24-0.98]). Metformin was related to lower odds of pathological p-tau when diabetes was uncontrolled, acting on t-tau and t-tau/Aβ42 ratio when it was concomitant with insulin, and patients had controlled diabetes. Lower odds of pathological levels of tau were observed when additional oral antidiabetic drugs were added among metformin users. iSGLT2 was associated with tau pathology. CONCLUSIONS The impact of antidiabetics on AD-related pathological biomarkers may depend on diabetes management.
Collapse
Affiliation(s)
- Gemma García-Lluch
- Research Group in Alzheimer Disease, Health Research Institute La Fe, Valencia, Spain
- Cathedra DeCo MICOF-CEU UCH, University Cardenal Herrera-CEU, Valencia, Spain
- Department of Pharmacy, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Anna Marseglia
- Department of Pharmacy, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Lucrecia Moreno Royo
- Cathedra DeCo MICOF-CEU UCH, University Cardenal Herrera-CEU, Valencia, Spain
- Department of Pharmacy, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Juan Pardo Albiach
- Embedded Systems and Artificial Intelligence Group, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Mar Garcia-Zamora
- Research Group in Alzheimer Disease, Health Research Institute La Fe, Valencia, Spain
- Cathedra DeCo MICOF-CEU UCH, University Cardenal Herrera-CEU, Valencia, Spain
| | - Miquel Baquero
- Research Group in Alzheimer Disease, Health Research Institute La Fe, Valencia, Spain
- Cathedra DeCo MICOF-CEU UCH, University Cardenal Herrera-CEU, Valencia, Spain
- Neurology Unit, University and Polytechnic Hospital La Fe, Valencia, Spain
| | - Carmen Peña-Bautista
- Research Group in Alzheimer Disease, Health Research Institute La Fe, Valencia, Spain
| | - Lourdes Álvarez
- Research Group in Alzheimer Disease, Health Research Institute La Fe, Valencia, Spain
| | - Eric Westman
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Consuelo Cháfer-Pericás
- Research Group in Alzheimer Disease, Health Research Institute La Fe, Valencia, Spain
- Cathedra DeCo MICOF-CEU UCH, University Cardenal Herrera-CEU, Valencia, Spain
| |
Collapse
|
7
|
Yin Q, Yang G, Su R, Bu J, Li Y, Zhang H, Zhang Y, Zhuang P. Zi Shen Wan Fang repaired blood-brain barrier integrity in diabetic cognitive impairment mice via preventing cerebrovascular cells senescence. Chin Med 2024; 19:169. [PMID: 39696612 DOI: 10.1186/s13020-024-01041-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Blood-brain barrier (BBB) integrity disruption is a key pathological link of diabetes-induced cognitive impairment (DCI), but the detailed mechanism of how the diabetic environment induces BBB integrity disruption is not fully understood. Our previous study found that Zi Shen Wan Fang (ZSWF), an optimized prescription consisting of Anemarrhenae Rhizoma (Anemarrhena asphodeloides Bge.), Phellodendri Chinensis Cortex (Phellodendron chinense Schneid.) and Cistanches Herba (Cistanche deserticola Y.C.Ma) has excellent efficacy in alleviating DCI, however, whether its mechanism is related to repairing BBB integrity remains unclear. This study aims to reveal the mechanism of BBB integrity destruction in DCI mice, and to elucidate the mechanism by which ZSWF repairs BBB integrity and improves cognitive function in DCI mice. METHODS Diabetic mouse model was established by feeding a 60% high-fat diet combined with a single intraperitoneal injection of 120 mg/kg streptozotocin (STZ). DCI mice were screened with morris water maze (MWM) after 8 weeks of sustained hyperglycemic stimulation. ZSWF was administered daily at doses of 9.36 and 18.72 g/kg for 8 weeks. Cognitive function was evaluated using MWM, blood-brain-barrier (BBB) integrity was tested using immunostaining and western blot, the underlying mechanisms were explored using single-cell RNA sequencing (scRNA-seq), validation experiments were performed with immunofluorescence analysis, and the potential active ingredients of ZSWF against cerebrovascular senescence were predicted using molecular docking. Moreover, cerebral microvascular endothelial cells were cultured, and the effects of mangiferin on the expression of p21 and Vcam1 were investigated by immunofluorescence staining and RT-qPCR. RESULTS ZSWF treatment significantly ameliorated cognitive function and repaired BBB integrity in DCI mice. Using scRNA-seq, we identified 14 brain cell types. In BBB constituent cells (endothelial cells and pericytes), we found that Cdkn1a and senescence-associated secretory phenotype (SASP) genes were significantly overexpressed in DCI mice, while ZSWF intervention significantly inhibited the expression of Cdkn1a and SASP genes in cerebrovascular cells of DCI mice. Moreover, we also found that the communication between brain endothelial cells and pericytes was decreased in DCI mice, while ZSWF significantly increased the communication between them, especially the expression of PDGFRβ in pericytes. Molecular docking results showed that mangiferin, the blood component of ZSWF, had a stronger affinity with the upstream proteins of p21. In vitro experiments showed that high glucose significantly increased the expression of p21 and Vcam1 in bEnd.3 cells, while mangiferin significantly inhibited the expression of p21 and Vcam1 induced by high glucose. CONCLUSION Our study reveals that ZSWF can ameliorate cognitive function in DCI mice by repairing BBB integrity, and the specific mechanism of which may be related to preventing cerebrovascular cells senescence, and mangiferin is its key active ingredient.
Collapse
Affiliation(s)
- Qingsheng Yin
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Genhui Yang
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Runtao Su
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jie Bu
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Ying Li
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Han Zhang
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China.
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Yanjun Zhang
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China.
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- Department of Integrated Rehabilitation, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China.
| | - Pengwei Zhuang
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Jinghai District, Tianjin, 301617, China.
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- Department of Integrated Rehabilitation, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China.
| |
Collapse
|
8
|
Schwartz SS, Herman ME, Tun MTH, Barone E, Butterfield DA. The double life of glucose metabolism: brain health, glycemic homeostasis, and your patients with type 2 diabetes. BMC Med 2024; 22:582. [PMID: 39696300 DOI: 10.1186/s12916-024-03763-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 11/11/2024] [Indexed: 12/20/2024] Open
Abstract
The maintenance of cognitive function is essential for quality of life and health outcomes in later years. Cognitive impairment, however, remains an undervalued long-term complication of type 2 diabetes by patients and providers alike. The burden of sustained hyperglycemia includes not only cognitive deficits but also the onset and progression of dementia-related conditions, including Alzheimer's disease (AD). Recent research has shown that the brain maintains an independent glucose "microsystem"-evolved to ensure the availability of fuel for brain neurons without interruption by transient hypoglycemia. When this milieu is perturbed, brain hyperglycemia, brain glucotoxicity, and brain insulin resistance can ensue and interfere with insulin signaling, a key pathway to cognitive function and neuronal integrity. This newly understood brain homeostatic system operates semi-autonomously from the systemic glucoregulatory apparatus. Large-scale clinical studies have shown that systemic dysglycemia is also strongly associated with poorer cognitive outcomes, which can be mitigated through appropriate clinical management of plasma glucose levels. Moreover, these studies demonstrated that glucose-lowering agents are not equally effective at preventing cognitive dysfunction. Glucagon-like peptide-1 (GLP-1) receptor analogs and sodium glucose cotransporter 2 inhibitors (SGLT2is) appear to afford the greatest protection; metformin and dipeptidyl peptidase 4 inhibitors (DPP-4is) also significantly improved cognitive outcomes. Sulfonylureas (SUs) and exogenous insulin, on the other hand, do not provide the same protection and may actually worsen cognitive outcomes. In the creation of a treatment plan, comorbid cognitive conditions should be considered. These efficacious treatments create a new gold standard of managing hyperglycemia-one which is consistent with the "complication-centric prescribing" mandates issued in type 2 diabetes treatment guidelines. The increasing longevity enjoyed by our populace places the onus on clinical care to play the "long game" in using targeted treatments for glucose control in patients with, or at risk for, cognitive decline to maintain cognitive wellness later in life. This article reviews critical emerging data for scientists and trialists and translates new enhancements in patient care for practitioners.
Collapse
Affiliation(s)
- Stanley S Schwartz
- University of Pennsylvania School of Medicine, 771 County Line Road, Villanova, PA, 19085, USA
| | - Mary E Herman
- Social Alchemy: Building Physician Competency Across the Globe, 5 Ave Sur #36, Antigua, Sacatepéquez, Guatemala.
| | - May Thet Hmu Tun
- Maimonides Medical Center, 4802 10th Ave, Brooklyn, NY, 11219, USA
| | - Eugenio Barone
- Sapienza University of Rome, Via Degli Equi 42, Scala A, Int. 5, 00185, Rome, Italy
| | - D Allan Butterfield
- Sanders-Brown Center On Aging, Department of Chemistry, University of Kentucky, 249 Chemistry-Physics Building, Lexington, KY, 40506-0055, USA
| |
Collapse
|
9
|
Tran J, Parekh S, Rockcole J, Wilson D, Parmar MS. Repurposing antidiabetic drugs for Alzheimer's disease: A review of preclinical and clinical evidence and overcoming challenges. Life Sci 2024; 355:123001. [PMID: 39173996 DOI: 10.1016/j.lfs.2024.123001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/13/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Repurposing antidiabetic drugs for the treatment of Alzheimer's disease (AD) has emerged as a promising therapeutic strategy. This review examines the potential of repurposing antidiabetic drugs for AD treatment, focusing on preclinical evidence, clinical trials, and observational studies. In addition, the review aims to explore challenges and opportunities in repurposing antidiabetic drugs for AD, emphasizing the importance of well-designed clinical trials that consider patient selection criteria, refined outcome measures, adverse effects, and combination therapies to enhance therapeutic efficacy. Preclinical evidence suggests that glucagon-like peptide-1 (GLP-1) analogs, dipeptidyl peptidase-4 (DPP4) inhibitors, metformin, thiazolidinediones, and sodium-glucose co-transporter-2 (SGLT2) inhibitors exhibit neuroprotective effects in AD preclinical models. In preclinical studies, antidiabetic drugs have demonstrated neuroprotective effects by reducing amyloid beta (Aβ) plaques, tau hyperphosphorylation, neuroinflammation, and cognitive impairment. Antidiabetic drug classes, notably GLP-1 analogs and SGLT2 inhibitors, and a reduced risk of dementia in patients with diabetes mellitus. While the evidence for DPP4 inhibitors is mixed, some studies suggest a potential protective effect. On the other hand, alpha-glucosidase inhibitors (AGIs) and sulfonylureas may potentially increase the risk, especially in those experiencing recurrent hypoglycemic events. Repurposing antidiabetic drugs for AD is a promising therapeutic strategy, but challenges such as disease heterogeneity, limited biomarkers, and benefits versus risk evaluation need to be addressed. Ongoing clinical trials in mild cognitive impairment (MCI) and early AD patients without diabetes will be crucial in determining the clinical efficacy and safety of the antidiabetic drugs, paving the way for potential treatments for AD.
Collapse
Affiliation(s)
- Jacky Tran
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA
| | - Sneh Parekh
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA
| | - Julia Rockcole
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA
| | - Danielle Wilson
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA
| | - Mayur S Parmar
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA.
| |
Collapse
|
10
|
Tang H, Donahoo WT, Svensson M, Shaaban CE, Smith G, Jaffee MS, Huang Y, Hu X, Lu Y, Salloum RG, DeKosky ST, Bian J, Guo J. Heterogeneous treatment effects of sodium-glucose cotransporter 2 inhibitors on risk of dementia in people with type 2 diabetes: A population-based cohort study. Alzheimers Dement 2024; 20:5528-5539. [PMID: 38958394 PMCID: PMC11350016 DOI: 10.1002/alz.14048] [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: 01/04/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 07/04/2024]
Abstract
INTRODUCTION Sodium-glucose cotransporter 2 (SGLT2) inhibitors exhibit potential benefits in reducing dementia risk, yet the optimal beneficiary subgroups remain uncertain. METHODS Individuals with type 2 diabetes (T2D) initiating either SGLT2 inhibitor or sulfonylurea were identified from OneFlorida+ Clinical Research Network (2016-2022). A doubly robust learning was deployed to estimate risk difference (RD) and 95% confidence interval (CI) of all-cause dementia. RESULTS Among 35,458 individuals with T2D, 1.8% in the SGLT2 inhibitor group and 4.7% in the sulfonylurea group developed all-cause dementia over a 3.2-year follow-up, yielding a lower risk for SGLT2 inhibitors (RD, -2.5%; 95% CI, -3.0% to -2.1%). Hispanic ethnicity and chronic kidney disease were identified as the two important variables to define four subgroups in which RD ranged from -4.3% (-5.5 to -3.2) to -0.9% (-1.9 to 0.2). DISCUSSION Compared to sulfonylureas, SGLT2 inhibitors were associated with a reduced risk of all-cause dementia, but the association varied among different subgroups. HIGHLIGHTS New users of sodium-glucose cotransporter 2 (SGLT2) inhibitors were significantly associated with a lower risk of all-cause dementia as compared to those of sulfonylureas. The association varied among different subgroups defined by Hispanic ethnicity and chronic kidney disease. A significantly lower risk of Alzheimer's disease and vascular dementia was observed among new users of SGLT2 inhibitors compared to those of sulfonylureas.
Collapse
Affiliation(s)
- Huilin Tang
- Department of Pharmaceutical Outcomes and PolicyUniversity of Florida College of PharmacyGainesvilleFloridaUSA
| | - William T. Donahoo
- Department of MedicineUniversity of Florida College of MedicineGainesvilleFloridaUSA
| | - Mikael Svensson
- Department of Pharmaceutical Outcomes and PolicyUniversity of Florida College of PharmacyGainesvilleFloridaUSA
- Center for Drug Evaluation and SafetyUniversity of FloridaGainesvilleFloridaUSA
| | - C. Elizabeth Shaaban
- Department of EpidemiologySchool of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
- Alzheimer's Disease Research CenterUniversity of PittsburghPennsylvaniaUSA
| | - Glenn Smith
- Department of Clinical and Health PsychologyCollege of Public Health and Health ProfessionsUniversity of FloridaGainesvilleFloridaUSA
- 1Florida Alzheimer's Disease Research Center (ADRC)University of FloridaGainesvilleFloridaUSA
| | - Michael S. Jaffee
- 1Florida Alzheimer's Disease Research Center (ADRC)University of FloridaGainesvilleFloridaUSA
- Department of Neurology and McKnight Brain InstituteCollege of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Yu Huang
- Department of Health Outcomes and Biomedical InformaticsCollege of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Xia Hu
- DATA Lab, Department of Computer ScienceRice UniversityHoustonTexasUSA
| | - Ying Lu
- Department of Pharmaceutical Outcomes and PolicyUniversity of Florida College of PharmacyGainesvilleFloridaUSA
| | - Ramzi G. Salloum
- Department of Health Outcomes and Biomedical InformaticsCollege of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Steven T. DeKosky
- 1Florida Alzheimer's Disease Research Center (ADRC)University of FloridaGainesvilleFloridaUSA
- Department of Neurology and McKnight Brain InstituteCollege of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Jiang Bian
- Department of Health Outcomes and Biomedical InformaticsCollege of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Jingchuan Guo
- Department of Pharmaceutical Outcomes and PolicyUniversity of Florida College of PharmacyGainesvilleFloridaUSA
- Center for Drug Evaluation and SafetyUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
11
|
Zamora MG, García-Lluch G, Moreno L, Pardo J, Pericas CC. Assessment of sodium-glucose cotransporter 2 inhibitors (SGLT2i) and other antidiabetic agents in Alzheimer's disease: A population-based study. Pharmacol Res 2024; 206:107295. [PMID: 38971270 DOI: 10.1016/j.phrs.2024.107295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/18/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
The lack of effective treatments for dementia has led to explore the potential of antidiabetic agents as a possible approach. This cross-sectional and population-based study aimed to investigate the relationship between each antidiabetic drug and their defined daily doses (DDDs) and the use of anti-Alzheimer's disease (AD) drugs in order to establish new possible hypotheses about the role of antidiabetic drugs in AD. For that purpose, a database containing information on medications prescribed to 233183 patients aged 50 years or older between 2018 and 2020 was used. DDDs were calculated according to the ATC/DDD index 2023. Statistical analyses, with logistic regression, were carried out to assess antidiabetic and anti-AD drugs consumption. A total of 91836 patients who were prescribed at least one antihypertensive, antidiabetic, or lipid-modifying agent were included in the study; specifically, 29260 patients were prescribed antidiabetic medication. Among the antidiabetic agents, glucagon-like peptide-1 analogs (GLP-1) DDDs were likely to have a positive association with anti-AD drugs in people aged between 70 and 80 years. Additionally, sodium-glucose cotransporter 2 inhibitors (SGLT2i) were prone to have a positive association with anti-AD drug usage across almost every age. However, insulin usage was associated with an increased usage of anti-AD agents. In conclusion, there is evidence suggesting a correlation between certain antidiabetic agents and dementia. Specifically, GLP-1 and SGLT2i might be associated with lower odds of anti-AD drugs usage, while insulins might be linked to higher odds of using anti-AD drugs.
Collapse
Affiliation(s)
- Mar Garcia Zamora
- Research Group in Alzheimer Disease. Instituto de Investigación Sanitaria La Fe, Valencia, Spain; Cathedra DeCo MICOF-CEU UCH, University Cardenal Herrera-CEU, CEU Universities, Valencia 46115, Spain
| | - Gemma García-Lluch
- Research Group in Alzheimer Disease. Instituto de Investigación Sanitaria La Fe, Valencia, Spain; Cathedra DeCo MICOF-CEU UCH, University Cardenal Herrera-CEU, CEU Universities, Valencia 46115, Spain
| | - Lucrecia Moreno
- Cathedra DeCo MICOF-CEU UCH, University Cardenal Herrera-CEU, CEU Universities, Valencia 46115, Spain; Department of Pharmacy, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia 46115, Spain
| | - Juan Pardo
- Embedded Systems and Artificial Intelligence Group, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia 46115, Spain.
| | - Consuelo Cháfer Pericas
- Research Group in Alzheimer Disease. Instituto de Investigación Sanitaria La Fe, Valencia, Spain.
| |
Collapse
|
12
|
Peng Y, Yao SY, Chen Q, Jin H, Du MQ, Xue YH, Liu S. True or false? Alzheimer's disease is type 3 diabetes: Evidences from bench to bedside. Ageing Res Rev 2024; 99:102383. [PMID: 38955264 DOI: 10.1016/j.arr.2024.102383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024]
Abstract
Globally, Alzheimer's disease (AD) is the most widespread chronic neurodegenerative disorder, leading to cognitive impairment, such as aphasia and agnosia, as well as mental symptoms, like behavioral abnormalities, that place a heavy psychological and financial burden on the families of the afflicted. Unfortunately, no particular medications exist to treat AD, as the current treatments only impede its progression.The link between AD and type 2 diabetes (T2D) has been increasingly revealed by research; the danger of developing both AD and T2D rises exponentially with age, with T2D being especially prone to AD. This has propelled researchers to investigate the mechanism(s) underlying this connection. A critical review of the relationship between insulin resistance, Aβ, oxidative stress, mitochondrial hypothesis, abnormal phosphorylation of Tau protein, inflammatory response, high blood glucose levels, neurotransmitters and signaling pathways, vascular issues in AD and diabetes, and the similarities between the two diseases, is presented in this review. Grasping the essential mechanisms behind this detrimental interaction may offer chances to devise successful therapeutic strategies.
Collapse
Affiliation(s)
- Yong Peng
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China; Department of Neurology, Affiliated Provincial Traditional Chinese Medical Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China.
| | - Shun-Yu Yao
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China; Department of Neurology, Affiliated Provincial Traditional Chinese Medical Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Quan Chen
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China; Department of Neurology, Affiliated Provincial Traditional Chinese Medical Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Hong Jin
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China; Department of Neurology, Affiliated Provincial Traditional Chinese Medical Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Miao-Qiao Du
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China; Department of Neurology, Affiliated Provincial Traditional Chinese Medical Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Ya-Hui Xue
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China; Department of Neurology, Affiliated Provincial Traditional Chinese Medical Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| | - Shu Liu
- Department of Neurology, Affiliated First Hospital of Hunan Traditional Chinese Medical College, Zhuzhou, Hunan, China; Department of Neurology, Affiliated Provincial Traditional Chinese Medical Hospital of Hunan University of Chinese Medicine, Zhuzhou, Hunan, China
| |
Collapse
|
13
|
Martirosian RA, Wiedner CD, Sanchez J, Mun KT, Marla K, Teran C, Thirion M, Liebeskind DS, McGrath ER, Zucker JM, Bernal R, Beiser AS, DeCarli C, Himali JJ, Seshadri S, Hinman JD. Association of Incident Stroke Risk With an IL-18-Centered Inflammatory Network Biomarker Composite. Stroke 2024; 55:1601-1608. [PMID: 38690658 DOI: 10.1161/strokeaha.123.044719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 03/20/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND A coordinated network of circulating inflammatory molecules centered on the pleotropic pro-atherogenic cytokine interleukin-18 (IL-18) is linked to cerebral small vessel disease. We sought to validate the association of this inflammatory biomarker network with incident stroke risk, cognitive impairment, and imaging metrics in a sample of the Framingham Offspring Cohort. METHODS Using available baseline measurements of serum levels of IL-18, GDF (growth and differentiation factor)-15, soluble form of receptor for advanced glycation end products, myeloperoxidase, and MCP-1 (monocyte chemoattractant protein-1) from Exam 7 of the Framingham Offspring Cohort (1998-2001), we constructed a population-normalized, equally weighted log-transformed mean Z-score value representing the average level of each serum analyte to create an inflammatory composite score (ICS5). Multivariable regression models were used to determine the association of ICS5 with incident stroke, brain magnetic resonance imaging features, and cognitive testing performance. RESULTS We found a significant association between ICS5 score and increased risk for incident all-cause stroke (hazard ratio, 1.48 [95% CI, 1.05-2.08]; P=0.024) and ischemic stroke (hazard ratio, 1.51 [95% CI, 1.03-2.21]; P=0.033) in the Exam 7 cohort of 2201 subjects (mean age 62±9 years; 54% female) aged 45+ years with an all-cause incident stroke rate of 6.1% (135/2201) and ischemic stroke rate of 4.9% (108/2201). ICS5 and its component serum markers are all associated with the Framingham Stroke Risk Profile score (β±SE, 0.19±0.02; P<0.0001). In addition, we found a significant inverse association of ICS5 with a global cognitive score, derived from a principal components analysis of the neuropsychological battery used in the Framingham cohort (-0.08±0.03; P=0.019). No association of ICS5 with magnetic resonance imaging metrics of cerebral small vessel disease was observed. CONCLUSIONS Circulating serum levels of inflammatory biomarkers centered on IL-18 are associated with an increased risk of stroke and cognitive impairment in the Framingham Offspring Cohort. Linking specific inflammatory pathways to cerebral small vessel disease may enhance individualized quantitative risk assessment for future stroke and vascular cognitive impairment.
Collapse
Affiliation(s)
- Richard A Martirosian
- David Geffen School of Medicine, University of California Los Angeles (R.A.M., J.S., K.T.M., K.M., C.T., M.T., D.S.L., J.D.H.)
| | - Crystal D Wiedner
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases (C.D.W., R.B., J.J.H., S.S.), University of Texas Health Science Center at San Antonio
| | - Jasmin Sanchez
- David Geffen School of Medicine, University of California Los Angeles (R.A.M., J.S., K.T.M., K.M., C.T., M.T., D.S.L., J.D.H.)
| | - Katherine T Mun
- David Geffen School of Medicine, University of California Los Angeles (R.A.M., J.S., K.T.M., K.M., C.T., M.T., D.S.L., J.D.H.)
| | - Kiran Marla
- David Geffen School of Medicine, University of California Los Angeles (R.A.M., J.S., K.T.M., K.M., C.T., M.T., D.S.L., J.D.H.)
| | - Cristina Teran
- David Geffen School of Medicine, University of California Los Angeles (R.A.M., J.S., K.T.M., K.M., C.T., M.T., D.S.L., J.D.H.)
| | - Marissa Thirion
- David Geffen School of Medicine, University of California Los Angeles (R.A.M., J.S., K.T.M., K.M., C.T., M.T., D.S.L., J.D.H.)
| | - David S Liebeskind
- David Geffen School of Medicine, University of California Los Angeles (R.A.M., J.S., K.T.M., K.M., C.T., M.T., D.S.L., J.D.H.)
| | - Emer R McGrath
- Framingham Heart Study, MA (E.R.M.G., J.M.Z., A.S.B., C.D.C., J.J.H., S.S.)
- HRB Clinical Research Facility, School of Medicine, University of Galway, Ireland (E.R.M.G.)
| | - Jared M Zucker
- Framingham Heart Study, MA (E.R.M.G., J.M.Z., A.S.B., C.D.C., J.J.H., S.S.)
| | - Rebecca Bernal
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases (C.D.W., R.B., J.J.H., S.S.), University of Texas Health Science Center at San Antonio
| | - Alexa S Beiser
- Framingham Heart Study, MA (E.R.M.G., J.M.Z., A.S.B., C.D.C., J.J.H., S.S.)
- Department of Neurology, Boston University School of Medicine, MA (A.S.B., J.J.H., S.S.)
- Department of Biostatistics, Boston University School of Public Health, MA (A.S.B., J.J.H.)
| | - Charles DeCarli
- Framingham Heart Study, MA (E.R.M.G., J.M.Z., A.S.B., C.D.C., J.J.H., S.S.)
- Department of Neurology, University of California Davis, Sacramento (C.D.C.)
| | - Jayandra J Himali
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases (C.D.W., R.B., J.J.H., S.S.), University of Texas Health Science Center at San Antonio
- Department of Population Health Sciences (J.J.H.), University of Texas Health Science Center at San Antonio
- Framingham Heart Study, MA (E.R.M.G., J.M.Z., A.S.B., C.D.C., J.J.H., S.S.)
- Department of Neurology, Boston University School of Medicine, MA (A.S.B., J.J.H., S.S.)
- Department of Biostatistics, Boston University School of Public Health, MA (A.S.B., J.J.H.)
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases (C.D.W., R.B., J.J.H., S.S.), University of Texas Health Science Center at San Antonio
- Framingham Heart Study, MA (E.R.M.G., J.M.Z., A.S.B., C.D.C., J.J.H., S.S.)
- Department of Neurology, Boston University School of Medicine, MA (A.S.B., J.J.H., S.S.)
| | - Jason D Hinman
- David Geffen School of Medicine, University of California Los Angeles (R.A.M., J.S., K.T.M., K.M., C.T., M.T., D.S.L., J.D.H.)
| |
Collapse
|
14
|
Cui W, Lv C, Geng P, Fu M, Zhou W, Xiong M, Li T. Novel targets and therapies of metformin in dementia: old drug, new insights. Front Pharmacol 2024; 15:1415740. [PMID: 38881878 PMCID: PMC11176471 DOI: 10.3389/fphar.2024.1415740] [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: 04/11/2024] [Accepted: 05/16/2024] [Indexed: 06/18/2024] Open
Abstract
Dementia is a devastating disorder characterized by progressive and persistent cognitive decline, imposing a heavy public health burden on the individual and society. Despite numerous efforts by researchers in the field of dementia, pharmacological treatments are limited to relieving symptoms and fail to prevent disease progression. Therefore, studies exploring novel therapeutics or repurposing classical drugs indicated for other diseases are urgently needed. Metformin, a first-line antihyperglycemic drug used to treat type 2 diabetes, has been shown to be beneficial in neurodegenerative diseases including dementia. This review discusses and evaluates the neuroprotective role of metformin in dementia, from the perspective of basic and clinical studies. Mechanistically, metformin has been shown to improve insulin resistance, reduce neuronal apoptosis, and decrease oxidative stress and neuroinflammation in the brain. Collectively, the current data presented here support the future potential of metformin as a potential therapeutic strategy for dementia. This study also inspires a new field for future translational studies and clinical research to discover novel therapeutic targets for dementia.
Collapse
Affiliation(s)
- Wenxing Cui
- College of Life Sciences, Northwest University, Xi’an, China
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - Chen Lv
- Hangzhou Simo Co., Ltd., Hangzhou, China
| | - Panling Geng
- College of Life Sciences, Northwest University, Xi’an, China
| | - Mingdi Fu
- College of Life Sciences, Northwest University, Xi’an, China
| | - Wenjing Zhou
- College of Life Sciences, Northwest University, Xi’an, China
| | - Mingxiang Xiong
- College of Life Sciences, Northwest University, Xi’an, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
15
|
Youn YJ, Kim S, Jeong HJ, Ah YM, Yu YM. Sodium-glucose cotransporter-2 inhibitors and their potential role in dementia onset and cognitive function in patients with diabetes mellitus: a systematic review and meta-analysis. Front Neuroendocrinol 2024; 73:101131. [PMID: 38367940 DOI: 10.1016/j.yfrne.2024.101131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 02/03/2024] [Accepted: 02/10/2024] [Indexed: 02/19/2024]
Abstract
This systematic review and meta-analysis aimed to determine the association between the use of sodium-glucose cotransporter 2 (SGLT-2) inhibitors and dementia onset as well as cognitive function in patients with diabetes mellitus. We comprehensively searched the MEDLINE, Embase, and CENTRAL databases to select relevant studies published up to August 2023. The use of SGLT-2 inhibitors significantly lowers dementia risk compared to SGLT-2i non-users (Hazard ratio: 0.68, 95 % CI: 0.50-0.92). Furthermore, our findings indicated a positive effect of SGLT-2 inhibitor use on cognitive function score improvement, as demonstrated by the standardized mean difference of 0.88 (95 % CI: 0.32-1.44), particularly among populations with mild cognitive impairment or dementia. This systematic review and meta-analysis indicate a potential role of SGLT-2 inhibitors in reducing the risk of dementia in patients with diabetes mellitus. These findings underscore the need for well-controlled large clinical trials and future research in this field.
Collapse
Affiliation(s)
- Yea Jin Youn
- Graduate Program of Industrial Pharmaceutical Science, Yonsei University, Incheon, Republic of Korea
| | - Seungyeon Kim
- College of Pharmacy, Dankook University, Cheonan, Republic of Korea
| | - Hyun-Jeong Jeong
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Young-Mi Ah
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea.
| | - Yun Mi Yu
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, Republic of Korea; Department of Pharmaceutical Medicine and Regulatory Sciences, Colleges of Medicine and Pharmacy, Yonsei University, Incheon, Republic of Korea.
| |
Collapse
|
16
|
Arvanitakis Z, Capuano AW, Tong H, Mehta RI, Anokye-Danso F, Bennett DA, Arnold SE, Ahima RS. Associations of Serum Insulin and Related Measures With Neuropathology and Cognition in Older Persons With and Without Diabetes. Ann Neurol 2024; 95:665-676. [PMID: 38379184 PMCID: PMC11023784 DOI: 10.1002/ana.26882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/23/2023] [Accepted: 01/26/2024] [Indexed: 02/22/2024]
Abstract
OBJECTIVE To examine associations of serum insulin and related measures with neuropathology and cognition in older persons. METHODS We studied 192 older persons (96 with diabetes and 96 without, matched by sex and balanced by age-at-death, education, and postmortem interval) from a community-based, clinical-pathologic study of aging, with annual evaluations including neuropsychological testing (summarized into global cognition and 5 cognitive domains) and postmortem autopsy. We assessed serum insulin, glucose, leptin, adiponectin, hemoglobin A1C, advanced glycation-end products (AGEs), and receptors for advanced glycation-end products, and calculated the Homeostasis Model Assessment of Insulin Resistance (HOMA-IR) and adiponectin-to-leptin ratio. Using adjusted regression analyses, we examined the associations of serum measures with neuropathology of cerebrovascular disease and Alzheimer's disease, and with the level of cognition proximate-to-death. RESULTS Higher HOMA-IR was associated with the presence of brain infarcts and specifically microinfarcts, and higher HOMA-IR and leptin were each associated with subcortical infarcts. Further, higher leptin levels and lower adiponectin-to-leptin ratios were associated with the presence of moderate-to-severe atherosclerosis. Serum insulin and related measures were not associated with the level of Alzheimer's disease pathology, as assessed by global, as well as amyloid burden or tau tangle density scores. Regarding cognitive outcomes, higher insulin and leptin levels, and lower adiponectin and receptors for advanced glycation-end products levels, respectively, were each associated with lower levels of global cognition. INTERPRETATION Peripheral insulin resistance indicated by HOMA-IR and related serum measures was associated with a greater burden of cerebrovascular neuropathology and lower cognition. ANN NEUROL 2024;95:665-676.
Collapse
Affiliation(s)
- Zoe Arvanitakis
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL
| | - Ana W Capuano
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL
| | - Han Tong
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL
| | - Rupal I Mehta
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL
| | - Frederick Anokye-Danso
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University School of Medicine, Baltimore, MD
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL
| | - Steven E Arnold
- Alzheimer's Clinical and Translational Research Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Rexford S Ahima
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
17
|
Alkabbani W, Maxwell CJ, Marrie RA, Tyas SL, Lega IC, Gamble JM. Insulin, hypoglycaemia and dementia: A causal mediation analysis showcasing challenges and potential opportunities. Diabetes Obes Metab 2024; 26:1120-1124. [PMID: 38012861 DOI: 10.1111/dom.15385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/10/2023] [Accepted: 11/12/2023] [Indexed: 11/29/2023]
Affiliation(s)
- Wajd Alkabbani
- School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada
| | - Colleen J Maxwell
- School of Pharmacy, University of Waterloo, Waterloo, Ontario, Canada
- School of Public Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - Ruth Ann Marrie
- Departments of Internal Medicine and Community Health Science, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Suzanne L Tyas
- School of Public Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - Iliana C Lega
- Women's College Research Institute (WCRI), Women's College Hospital, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
18
|
Pase MP, Himali JJ, Puerta R, Beiser AS, Gonzales MM, Satizabal CL, Yang Q, Aparicio HJ, Kojis DJ, Decarli CS, Lopez OL, Longstreth W, Gudnason V, Mosley TH, Bis JC, Fohner A, Psaty BM, Boada M, García-González P, Valero S, Marquié M, Tracy R, Launer LJ, Ruiz A, Fornage M, Seshadri S. Association of Plasma YKL-40 With MRI, CSF, and Cognitive Markers of Brain Health and Dementia. Neurology 2024; 102:e208075. [PMID: 38290090 PMCID: PMC11383876 DOI: 10.1212/wnl.0000000000208075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/07/2023] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Higher YKL-40 levels in the CSF are a known biomarker of brain inflammation. We explored the utility of plasma YKL-40 as a biomarker for accelerated brain aging and dementia risk. METHODS We performed cross-sectional and prospective analyses of 4 community-based cohorts in the United States or Europe: the Age, Gene/Environment Susceptibility-Reykjavik Study, Atherosclerosis Risk in the Communities study, Coronary Artery Risk Development in Young Adults study, and Framingham Heart Study (FHS). YKL-40 was measured from stored plasma by a single laboratory using Mesoscale Discovery with levels log transformed and standardized within each cohort. Outcomes included MRI total brain volume, hippocampal volume, and white matter hyperintensity volume (WMHV) as a percentage of intracranial volume, a general cognitive composite derived from neuropsychological testing (SD units [SDU]), and the risk of incident dementia. We sought to replicate associations with dementia in the clinic-based ACE csf cohort, which also had YKL-40 measured from the CSF. RESULTS Meta-analyses of MRI outcomes included 6,558 dementia-free participants, and for analysis of cognition, 6,670. The blood draw preceded MRI/cognitive assessment by up to 10.6 years across cohorts. The mean ages ranged from 50 to 76 years, with 39%-48% male individuals. In random-effects meta-analysis of study estimates, each SDU increase in log-transformed YKL-40 levels was associated with smaller total brain volume (β = -0.33; 95% CI -0.45 to -0.22; p < 0.0001) and poorer cognition (β = -0.04; 95% CI -0.07 to -0.02; p < 0.01), following adjustments for demographic variables. YKL-40 levels did not associate with hippocampal volume or WMHV. In the FHS, each SDU increase in log YKL-40 levels was associated with a 64% increase in incident dementia risk over a median of 5.8 years of follow-up, following adjustments for demographic variables (hazard ratio 1.64; 95% CI 1.25-2.16; p < 0.001). In the ACE csf cohort, plasma and CSF YKL-40 were correlated (r = 0.31), and both were associated with conversion from mild cognitive impairment to dementia, independent of amyloid, tau, and neurodegeneration status. DISCUSSION Higher plasma YKL-40 levels were associated with lower brain volume, poorer cognition, and incident dementia. Plasma YKL-40 may be useful for studying the association of inflammation and its treatment on dementia risk.
Collapse
Affiliation(s)
- Matthew P Pase
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Jayandra J Himali
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Raquel Puerta
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Alexa S Beiser
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Mitzi M Gonzales
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Claudia L Satizabal
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Qiong Yang
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Hugo J Aparicio
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Daniel J Kojis
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Charles S Decarli
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Oscar L Lopez
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Will Longstreth
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Vilmundur Gudnason
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Thomas H Mosley
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Joshua C Bis
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Alison Fohner
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Bruce M Psaty
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Mercè Boada
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Pablo García-González
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Sergi Valero
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Marta Marquié
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Russell Tracy
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Lenore J Launer
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Agustín Ruiz
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Myriam Fornage
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| | - Sudha Seshadri
- From the Turner Institute for Brain and Mental Health (M.P.P.), Monash University, Australia; Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases (J.J.H., M.M.G.), University of Texas Health Sciences Center, San Antonio; ACE Alzheimer Center (R.P., M.B., P.G.-G., S.V., M.M., A.R.), Barcelona, Spain; Boston University School of Public Health (A.S.B., D.J.K.), MA; University of Texas Health Sciences Center (C.L.S., S.S.), San Antonio; Department of Neurology (Q.Y., H.J.A.), Boston University School of Medicine, MA; Department of Neurology (C.S.D.), School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California at Davis; Department of Neurology (O.L.L.), School of Medicine, University of Pittsburgh, PA; University of Washington (W.L., B.M.P.), Seattle; Faculty of Medicine (V.G.), University of Iceland, Reykjavík; University of Mississippi Medical Center (T.H.M.), The MIND Center, Jackson; Cardiovascular Health Research Unit (J.C.B.), Department of Medicine, and Department of Epidemiology (A.F.), University of Washington, Seattle; University of Vermont (R.T.), Burlington; Laboratory of Epidemiology and Population Sciences (L.J.L.), National Institute on Aging, NIH, Bethesda, MD; and University of Texas Health Science Center (M.F.), Houston. Matthew P. Pase is currently at the School of Psychological Sciences and the Turner Institute for Brain and Mental Health, Monash University, Australia
| |
Collapse
|
19
|
Kim HK, Lee W, Ryu IH, Kim JK, Kim H, Yoo TK. Association between metformin use and the risk of developing open-angle glaucoma among patients with diabetes: a retrospective cohort study and meta-analysis. Int Ophthalmol 2024; 44:6. [PMID: 38316664 DOI: 10.1007/s10792-024-02945-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 12/07/2023] [Indexed: 02/07/2024]
Abstract
PURPOSE Recent studies examining the neuroprotective effects of metformin on open-angle glaucoma (OAG) have failed to provide consistent results. In this study, we investigated the association between metformin use and OAG. METHODS Data were obtained from a sample cohort of the Korean National Health Insurance database. Patients diagnosed with type-2 diabetes (T2DM) between 2004 and 2013 were included. We performed propensity score-matched analysis in a matched cohort (N = 20,646). The risk of the newly developed OAG was estimated using a Cox proportional hazards model. Including the present study, the meta-analysis included five studies to calculate the pooled risk for OAG based on metformin use. RESULTS In the adjusted model, the analysis revealed no statistical association between metformin use and OAG incidence (hazard ratio [HR] 1.05; 95% confidence interval [CI] 0.79-1.40; P = 0.738). The highest tercile of metformin use demonstrated no statistical significance (HR 0.93 [95% CI 0.63-1.37]; P = 0.703). No significant dose-dependent association was observed between the cumulative dose and incidence of OAG (P-value for trend = 0.336). In a meta-analysis of four published articles and the present study, the common-effects and random-effects models indicated conflicting results in terms of significance. The random effects model demonstrated no significant association (pooled risk ratio 0.53; 95% CI 0.24-1.19; P = 0.123). CONCLUSION We found no significant association between metformin use and OAG incidence in patients with T2DM in this population-based cohort study and meta-analysis. Further studies are needed to investigate the association between metformin use and the risk of OAG among patients with T2DM.
Collapse
Affiliation(s)
- Hong Kyu Kim
- Department of Ophthalmology, Dankook University Hospital, Dankook University College of Medicine, Cheonan, Republic of Korea
| | - Wanhyung Lee
- Department of Preventive Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Ik Hee Ryu
- Department of Ophthalmology, B&VIIT Eye Center, Seoul, Republic of Korea
| | - Jin Kuk Kim
- Department of Ophthalmology, B&VIIT Eye Center, Seoul, Republic of Korea
| | | | - Tae Keun Yoo
- Department of Ophthalmology, B&VIIT Eye Center, Seoul, Republic of Korea.
- Department of Refractive Surgery; VISUWORKS, B&VIIT Eye Center, B2 GT Tower, 1317-23, Seocho-Dong, Seocho-Gu, Seoul, Republic of Korea.
| |
Collapse
|
20
|
Tahmi M, Benitez R, Luchsinger JA. Metformin as a Potential Prevention Strategy for Alzheimer's Disease and Alzheimer's Disease Related Dementias. J Alzheimers Dis 2024; 101:S345-S356. [PMID: 39422959 DOI: 10.3233/jad-240495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Background Metformin is a safe and effective medication for type 2 diabetes (T2D) that has been proposed to decrease the risk of aging related disorders including Alzheimer's disease (AD) and Alzheimer's disease related disorders(ADRD). Objective This review seeks to summarize findings from studies examining the association of metformin with AD/ADRD related outcomes. Methods This is a narrative review of human studies, including observational studies and clinical trials, examining the association of metformin with cognitive and brain outcomes. We used PubMed as the main database for our literature search with a focus on English language human studies including observational studies and clinical trials. We prioritized studies published from 2013 until February 15, 2024. Results Observational human studies are conflicting, but those with better study designs suggest that metformin use in persons with T2D is associated with a lower risk of dementia. However, these observational studies are limited by the use of administrative data to ascertain metformin use and/or cognitive outcomes. There are few clinical trials in persons without T2D that have small sample sizes and short durations but suggest that metformin could prevent AD/ADRD. There are ongoing studies including large clinical trials with long duration that are testing the effect of metformin on AD/ADRD outcomes in persons without T2D at risk for dementia. Conclusions Clinical trial results are needed to establish the effect of metformin on the risk of AD and ADRD.
Collapse
Affiliation(s)
- Mouna Tahmi
- Department of Neurology, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Richard Benitez
- Departments of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - José A Luchsinger
- Departments of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
- Department of Epidemiology, Joseph P. Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
21
|
Wu CY, Wang C, Saskin R, Shah BR, Kapral MK, Lanctôt KL, Herrmann N, Cogo-Moreira H, MacIntosh BJ, Edwards JD, Swardfager W. No association between metformin initiation and incident dementia in older adults newly diagnosed with diabetes. J Intern Med 2024; 295:68-78. [PMID: 37747779 DOI: 10.1111/joim.13723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
BACKGROUND Metformin has been suggested to reduce dementia risk; however, most epidemiologic studies have been limited by immortal time bias or confounding due to disease severity. OBJECTIVES To investigate the association of metformin initiation with incident dementia using strategies that mitigate these important sources of bias. METHODS Residents of Ontario, Canada ≥66 years newly diagnosed with diabetes from January 1, 2008 to December 31, 2017 entered this retrospective population-based cohort. To consider the indication for metformin monotherapy initiation, people with hemoglobin A1c of 6.5%-8.0% and estimated glomerular filtration rate ≥45 mL/min/1.73 m2 were selected. Using the landmark method to address immortal time bias, exposure was grouped into "metformin monotherapy initiation within 180 days after new diabetes diagnosis" or "no glucose-lowering medications within 180 days." To address disease latency, 1-year lag time was applied to the end of the 180-day landmark period. Incident dementia was defined using a validated algorithm for Alzheimer's disease and related dementias. Adjusted hazard ratios (aHR) and confidence intervals (CIs) were estimated from propensity-score weighted Cox proportional hazard models. RESULTS Over mean follow-up of 6.77 years from cohort entry, metformin initiation within 180 days after new diabetes diagnosis (N = 12,331; 978 events; 65,762 person-years) showed no association with dementia risk (aHR [95% CI] = 1.05 [0.96-1.15]), compared to delayed or no glucose-lowering medication initiation (N = 22,369; 1768 events; 117,415 person-years). CONCLUSION Early metformin initiation was not associated with incident dementia in older adults newly diagnosed with diabetes. The utility of metformin to prevent dementia was not supported.
Collapse
Affiliation(s)
- Che-Yuan Wu
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
- Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | | | | | - Baiju R Shah
- ICES, Toronto, Ontario, Canada
- Divisions of Endocrinology and Obstetric Medicine, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Moira K Kapral
- ICES, Toronto, Ontario, Canada
- Institute for Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada
- Division of General Internal Medicine, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Krista L Lanctôt
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
- Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Psychiatry, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- KITE University Health Network Toronto Rehabilitation Institute, Toronto, Ontario, Canada
| | - Nathan Herrmann
- Department of Psychiatry, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Hugo Cogo-Moreira
- Faculty of Education, ICT, and Learning, Østfold University College, Halden, Norway
| | - Bradley J MacIntosh
- Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Radiology and Nuclear Medicine, Computational Radiology & Artificial Intelligence (CRAI), Oslo University Hospital, Oslo, Norway
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Jodi D Edwards
- University of Ottawa Heart Institute, University of Ottawa, Ottawa, Ontario, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
- ICES, Ottawa, Ontario, Canada
| | - Walter Swardfager
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
- Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- KITE University Health Network Toronto Rehabilitation Institute, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Xie Y, Wang J, Jiang J, Liu F, Zhang Y. Do oral antidiabetic medications alter the risk of Parkinson's disease? An updated systematic review and meta-analysis. Neurol Sci 2023; 44:4193-4203. [PMID: 37500992 DOI: 10.1007/s10072-023-06965-9] [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] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/14/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND Diabetes mellitus is a known risk factor for Parkinson's disease (PD), but does this risk vary with antidiabetic medications is still unclear. This meta-analysis aims to compile evidence from the literature to assess the risk of idiopathic PD with various oral antidiabetic medications. METHODS Databases PubMed, CENTRAL, Scopus, Web of Science, and Embase were searched till 5th April 2023. Adjusted outcomes were pooled to generate a hazard ratio (HR) on the risk of PD with different antidiabetic medications. RESULTS Fifteen studies with 2,910,405 diabetic patients were eligible. Pooled analysis failed to show any significant difference in the risk of PD among users of metformin (HR: 1.05 95% CI: 0.91, 1.22 I2 = 81%), glitazones (HR: 0.84 95% CI: 0.68, 1.05 I2 = 91%), glucagon-like peptide-1 agonists (HR: 0.63 95% CI: 0.26, 1.55 I2 = 33%), and sulfonylureas (HR: 1.13 95% CI: 0.96, 1.32 I2 = 76%). However, a meta-analysis of four studies showed that dipeptidyl peptidase-4 inhibitor use was associated with reduced risk of PD in diabetics (HR: 0.69 95% CI: 0.56, 0.86 I2 = 46%). Insufficient data was available on sodium-glucose cotransporter-2 inhibitors, α-glucosidase inhibitors, and glinides. CONCLUSIONS Limited retrospective evidence indicates that DPP4i may reduce the risk of idiopathic PD in diabetics. Metformin, sulfonylureas, glucagon-like peptide-1 agonists, and glitazones were not associated with any change in the risk of PD. Further studies taking into confounding factors and using a common comparator group are needed to strengthen present evidence.
Collapse
Affiliation(s)
- Yanna Xie
- Affiliated Hospital of Shaoxing University, NO. 999, Zhongxing South Road, Shaoxing City, Zhejiang Province, China
| | - Jialiang Wang
- Affiliated Hospital of Shaoxing University, NO. 999, Zhongxing South Road, Shaoxing City, Zhejiang Province, China
| | - Jie Jiang
- Affiliated Hospital of Shaoxing University, NO. 999, Zhongxing South Road, Shaoxing City, Zhejiang Province, China
| | - Furong Liu
- Affiliated Hospital of Shaoxing University, NO. 999, Zhongxing South Road, Shaoxing City, Zhejiang Province, China
| | - Yifang Zhang
- Affiliated Hospital of Shaoxing University, NO. 999, Zhongxing South Road, Shaoxing City, Zhejiang Province, China.
| |
Collapse
|
23
|
Slouha E, Ibrahim F, Rezazadah A, Esposito S, Clunes LA, Kollias TF. Anti-diabetics and the Prevention of Dementia: A Systematic Review. Cureus 2023; 15:e49515. [PMID: 38152822 PMCID: PMC10752751 DOI: 10.7759/cureus.49515] [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] [Accepted: 11/26/2023] [Indexed: 12/29/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a worldwide epidemic that is only increasing as the years progress, and as of 2019, affecting over 37 million. T2DM is a chronic condition caused by reduced insulin secretion and increased insulin resistance. Due to insulin not operating at optimal conditions, blood glucose rises and remains high, thus disturbing metabolic hemostasis. Many complications can arise from T2DM, such as coronary vascular disease, kidney damage, eye damage, and, quite significantly, dementia. It is theorized that dementia from T2DM stems from the fact that the brain is susceptible to hyperglycemic conditions, which are promoted by the increase in insulin resistance of target cells in the central nervous system. This directly affects cognitive processes and memory, which correlates to decreased temporal and front lobes volume. The risk of diabetic complications can be minimized with therapeutic interventions such as oral-antidiabetic (OAD) agents and insulin. Several OADs are on the market, but the first-line agent is metformin, a biguanide that decreases glucose production and increases insulin sensitivity. This paper aims to determine if currently prescribed OADs can help slow cognitive decline and reduce the risk and incidence of dementia as a complication of T2DM. Studies found that, for the most part, all OADs except sulfonylureas (SU) significantly slowed the decline of cognitive function and reduced the risk and incidence of dementia. SU's were shown to increase the risk of dementia in most studies. Of all the OADs, thiazolidinediones may be the most beneficial drug class for reducing the risk of dementia in T2DM patients. Future research should focus on whether early intervention with specific classes of OADs can not only improve glycemic control, leading to decreased hyperglycemia but also prevent the build-up of damaged brain tissue and help to reduce the risk and incidence of dementia in patients with T2DM.
Collapse
Affiliation(s)
- Ethan Slouha
- Anatomical Sciences, St. George's University School of Medicine, True Blue, GRD
| | - Fadi Ibrahim
- Pharmacology, St. George's University School of Medicine, True Blue, GRD
| | - Atbeen Rezazadah
- Pharmacology, St. George's University School of Medicine, True Blue, GRD
| | - Sarah Esposito
- Pharmacology, St. George's University School of Medicine, True Blue, GRD
| | - Lucy A Clunes
- Pharmacology, St. George's University, St George's, GRD
| | - Theofanis F Kollias
- Microbiology, Immunology and Pharmacology, St. George's University School of Medicine, True Blue, GRD
| |
Collapse
|
24
|
Zhou C, Dong C, Wang Q, Fu C, Xie Z, Hao W, Sun H, Zhu D. Healthy lifestyle and all-cause and cause-specific dementia in individuals with type 2 diabetes and the roles of diabetes duration and insulin use in UK Biobank cohort. Diabetes Obes Metab 2023; 25:3202-3211. [PMID: 37435782 DOI: 10.1111/dom.15215] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/13/2023]
Abstract
AIMS To examine the effect of a healthy lifestyle score derived from seven lifestyle factors recommended by the diabetes management guidelines on all-cause and cause-specific dementia in individuals with type 2 diabetes mellitus (T2DM), and how diabetes duration and insulin use status modify their association. MATERIALS AND METHODS This study analysed data of 459 840 participants from the UK Biobank. We used Cox proportional hazards models to estimate hazard ratios (HRs) and 95% confidence intervals for the association of an overall healthy lifestyle score with all-cause and cause-specific dementia of Alzheimer's disease, vascular dementia and non-Alzheimer non-vascular dementia. RESULTS Using diabetes-free participants who scored 5-7 as the reference group, in diabetes-free participants, we observed higher healthy lifestyle score was related to lower risk of all-cause and cause-specific dementia. However, in people with T2DM, those scored 2-3, 4 and 5-7 all had around the two-time risk of all-cause dementia (HR: 2.20-2.36), while those scored 0-1 had over a three-time risk (HR: 3.14, 95% confidence interval 2.34-4.21). A dose-response trend was observed with vascular dementia (each 2-point increase: 0.75, 0.61-0.93) and no significant association with Alzheimer's disease (0.95, 0.77-1.16). The reduced risk of all-cause and cause-specific dementia with higher lifestyle score was observed in patients with a diabetes duration less than 10 years, or in patients with no insulin use. CONCLUSION In people with T2DM, higher healthy lifestyle score was associated with lower risk of all-cause dementia. Diabetes duration and insulin use moderated the association between healthy lifestyle score and risk of dementia.
Collapse
Affiliation(s)
- Chunmiao Zhou
- Department of Epidemiology, School of Public Halth, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Caiyun Dong
- Department of Epidemiology, School of Public Halth, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qi Wang
- Centre for Health Management and Policy Research, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- NHC Key Lab of Health Economics and Policy Research (Shandong University), Jinan, China
| | - Chunying Fu
- Department of Epidemiology, School of Public Halth, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ziwei Xie
- Department of Epidemiology, School of Public Halth, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenting Hao
- Centre for Health Management and Policy Research, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- NHC Key Lab of Health Economics and Policy Research (Shandong University), Jinan, China
| | - Huizi Sun
- Department of Epidemiology, School of Public Halth, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dongshan Zhu
- Department of Epidemiology, School of Public Halth, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
25
|
Wu CY, Iskander C, Wang C, Xiong LY, Shah BR, Edwards JD, Kapral MK, Herrmann N, Lanctôt KL, Masellis M, Swartz RH, Cogo-Moreira H, MacIntosh BJ, Rabin JS, Black SE, Saskin R, Swardfager W. Association of sulfonylureas with the risk of dementia: A population-based cohort study. J Am Geriatr Soc 2023; 71:3059-3070. [PMID: 37218376 DOI: 10.1111/jgs.18397] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 03/09/2023] [Accepted: 04/20/2023] [Indexed: 05/24/2023]
Abstract
BACKGROUND Sulfonylureas are oral glucose-lowering medications positioned as a second-line therapy for type 2 diabetes. Evidence relating them to cognitive decline has been mixed. The objective was to determine whether sulfonylurea use was associated with a differential risk of dementia compared with dipeptidyl peptidase-4 (DPP4) inhibitor use. METHODS Using administrative data from residents in Ontario, Canada, adults aged ≥66 years who were new users of a sulfonylurea or a DPP4 inhibitor from June 14, 2011, to March 31, 2021 entered this population-based retrospective cohort study. Dementia was ascertained using a validated algorithm for Alzheimer's disease and related dementias. Propensity-score weighted Cox proportional hazards models were used to obtain adjusted hazard ratios (aHR) and confidence intervals (CI) for time to incident dementia. The observation window started at 1 year after cohort entry to mitigate protopathic bias due to delayed diagnosis. The primary analysis used an intention-to-treat exposure definition. A separate propensity-score weighted analysis was conducted to explore within-class differences in dementia risk among sulfonylurea new users selected from the primary cohort. RESULTS Among 107,806 DPP4 inhibitor new users and 37,030 sulfonylurea new users, sulfonylureas compared with DPP4 inhibitors were associated with a higher risk of dementia (18.4/1000 person-years; aHR [95% CI] = 1.09 [1.04-1.15]) over a mean follow-up of 4.82 years from cohort entry. Glyburide compared to gliclazide exhibited a higher dementia risk (aHR [95% CI] = 1.17 [1.03-1.32]). CONCLUSION New use of a sulfonylurea especially glyburide was associated with a higher dementia risk compared with new use of a DPP4 inhibitor in older adults with diabetes.
Collapse
Affiliation(s)
- Che-Yuan Wu
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
- Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | | | | | - Lisa Y Xiong
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
- Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Baiju R Shah
- ICES, Toronto, Ontario, Canada
- Divisions of Endocrinology and Obstetric Medicine, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Jodi D Edwards
- University of Ottawa Heart Institute, University of Ottawa, Ottawa, Ontario, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
- ICES, Ottawa, Ontario, Canada
| | - Moira K Kapral
- ICES, Toronto, Ontario, Canada
- Institute for Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada
- Division of General Internal Medicine, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Nathan Herrmann
- Department of Psychiatry, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Krista L Lanctôt
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
- Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Psychiatry, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- KITE University Health Network Toronto Rehabilitation Institute, Toronto, Ontario, Canada
- Toronto Dementia Research Alliance, Toronto, Ontario, Canada
| | - Mario Masellis
- Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Richard H Swartz
- Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- ICES, Toronto, Ontario, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Hugo Cogo-Moreira
- Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Faculty of Education, ICT, and Learning, Østfold University College, Halden, Norway
| | - Bradley J MacIntosh
- Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Computational Radiology & Artificial Intelligence (CRAI), Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Jennifer S Rabin
- Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
- Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Rehabilitation Sciences Institute, University of Toronto, Toronto, Ontario, Canada
| | - Sandra E Black
- Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Toronto Dementia Research Alliance, Toronto, Ontario, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | | | - Walter Swardfager
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
- Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- KITE University Health Network Toronto Rehabilitation Institute, Toronto, Ontario, Canada
| |
Collapse
|
26
|
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
|
27
|
Husain KH, Sarhan SF, AlKhalifa HKAA, Buhasan A, Moin ASM, Butler AE. Dementia in Diabetes: The Role of Hypoglycemia. Int J Mol Sci 2023; 24:9846. [PMID: 37372995 DOI: 10.3390/ijms24129846] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Hypoglycemia, a common consequence of diabetes treatment, is associated with severe morbidity and mortality and has become a major barrier to intensifying antidiabetic therapy. Severe hypoglycemia, defined as abnormally low blood glucose requiring the assistance of another person, is associated with seizures and comas, but even mild hypoglycemia can cause troubling symptoms such as anxiety, palpitations, and confusion. Dementia generally refers to the loss of memory, language, problem-solving, and other cognitive functions, which can interfere with daily life, and there is growing evidence that diabetes is associated with an increased risk of both vascular and non-vascular dementia. Neuroglycopenia resulting from a hypoglycemic episode in diabetic patients can lead to the degeneration of brain cells, with a resultant cognitive decline, leading to dementia. In light of new evidence, a deeper understating of the relationship between hypoglycemia and dementia can help to inform and guide preventative strategies. In this review, we discuss the epidemiology of dementia among patients with diabetes, and the emerging mechanisms thought to underlie the association between hypoglycemia and dementia. Furthermore, we discuss the risks of various pharmacological therapies, emerging therapies to combat hypoglycemia-induced dementia, as well as risk minimization strategies.
Collapse
Affiliation(s)
- Khaled Hameed Husain
- School of Medicine, Royal College of Surgeons in Ireland, Busaiteen, Adliya 15503, Bahrain
| | - Saud Faisal Sarhan
- School of Medicine, Royal College of Surgeons in Ireland, Busaiteen, Adliya 15503, Bahrain
| | | | - Asal Buhasan
- School of Medicine, Royal College of Surgeons in Ireland, Busaiteen, Adliya 15503, Bahrain
| | - Abu Saleh Md Moin
- Research Department, Royal College of Surgeons in Ireland, Busaiteen, Adliya 15503, Bahrain
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Busaiteen, Adliya 15503, Bahrain
| |
Collapse
|
28
|
Zhou C, Dong C, Xie Z, Hao W, Fu C, Sun H, Zhu D. Sex-specific associations between diabetes and dementia: the role of age at onset of disease, insulin use and complications. Biol Sex Differ 2023; 14:9. [PMID: 36804018 PMCID: PMC9940390 DOI: 10.1186/s13293-023-00491-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 02/07/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND Whether the association of type 2 diabetes (T2DM) with dementia was differed by sex remains unclear, and the roles of age at onset of disease, insulin use and diabetes' complications in their association are unknown. METHODS This study analyzed data of 447 931 participants from the UK Biobank. We used Cox proportional hazards models to estimate sex-specific hazard ratios (HRs) and 95% confidence intervals (CI), and women-to-men ratio of HRs (RHR) for the association between T2DM and incident dementia [all-cause dementia, Alzheimer's disease (AD), and vascular dementia (VD)]. The roles of age at onset of disease, insulin use and diabetes' complications in their association were also analyzed. RESULTS Compared to people with no diabetes at all, people with T2DM had increased risk of all-cause dementia (HR 2.85, 95% CI 2.56-3.17). The HRs between T2DM and AD were higher in women than men, with an RHR (95%CI) of 1.56 (1.20, 2.02). There was a trend that people who experienced T2DM before age 55 had higher risk of VD than those who had T2DM after age 55. In addition, there was a trend that T2DM had higher effect on VD that occurred before age 75 years than events that occurred after age 75. Patients with T2DM using insulin had higher risk of all-cause dementia than those without insulin, with an RHR (95%CI) of 1.54 (1.00-2.37). People with complications had doubled risk of all-cause dementia, AD and VD. CONCLUSIONS Adopting a sex-sensitive strategy to address the risk of dementia in patients with T2DM is instrumental for a precision medicine approach. Meanwhile, it is warranted to consider patients' age at onset of T2DM, insulin use status and complications conditions.
Collapse
Affiliation(s)
- Chunmiao Zhou
- grid.27255.370000 0004 1761 1174Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, 250012 Shandong China
| | - Caiyun Dong
- grid.27255.370000 0004 1761 1174Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, 250012 Shandong China
| | - Ziwei Xie
- grid.27255.370000 0004 1761 1174Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, 250012 Shandong China
| | - Wenting Hao
- grid.27255.370000 0004 1761 1174Centre for Health Management and Policy Research, School of Public Health, , Cheeloo College of Medicine, Shandong University, Jinan, 250012 China ,grid.27255.370000 0004 1761 1174NHC Key Lab of Health Economics and Policy Research (Shandong University), Jinan, 250012 China
| | - Chunying Fu
- grid.27255.370000 0004 1761 1174Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, 250012 Shandong China
| | - Huizi Sun
- grid.27255.370000 0004 1761 1174Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, 250012 Shandong China
| | - Dongshan Zhu
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, Shandong, China.
| |
Collapse
|
29
|
Abdelhafiz AH. Effects of hypoglycaemic therapy on frailty: a multi-dimensional perspective. Expert Rev Endocrinol Metab 2023; 18:53-65. [PMID: 36650694 DOI: 10.1080/17446651.2023.2168644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
INTRODUCTION The prevalence of diabetes is increasing in older people. With increasing age, frailty emerges as a new complication leading to disability. Frailty does not only include physical dysfunction but also involves negative impact on cognition and mood. Triad of impairments (TOI) is a new concept that includes physical frailty, dementia and depression to reflect the wider spectrum of frailty. AREAS COVERED Little is known about effects of hypoglycaemic agents on frailty syndrome. A literature search was performed on studies, which reported effects of hypoglycaemic agents on the component of the TOI. EXPERT OPINION It appears that most hypoglycaemic agents have some effects on frailty, although the results of clinical studies are inconsistent. Metformin seems to have a consistent and a positive effect on physical frailty. Its effects on cognitive function, however, are inconclusive but tend to be positive. Metformin appeared to improve depressive symptoms. Other agents such as incretins, thiazolidinediones, and sodium glucose transporter-2 inhibitors have some positive effects on cognition and depression. Sulfonylureas, glinides, or insulin have either negative or neutral effects on TOI components. The negative effects of insulin could be partially explained by the negative psychological factors and the frequent episodes of hypoglycemia associated with such therapy.
Collapse
Affiliation(s)
- Ahmed H Abdelhafiz
- Department of Geriatric Medicine, Rotherham General Hospital, Moorgate Road, Rotherham, UK
| |
Collapse
|
30
|
Sajadi E, Sajedianfard J, Hosseinzadeh S, Taherianfard M. Effect of insulin and cinnamon extract on spatial memory and gene expression of GLUT1, 3, and 4 in streptozotocin-induced Alzheimer's model in rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:680-687. [PMID: 37275760 PMCID: PMC10237167 DOI: 10.22038/ijbms.2023.68568.14957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 03/18/2023] [Indexed: 06/07/2023]
Abstract
Objectives Since diminished hippocampal insulin signaling leads to memory impairment, insulin resistance and hyperinsulinemia are probably associated with Alzheimer's disease (AD). The effect of intracerebroventricular injection of insulin (Ins) and oral cinnamon extract (Cinn) on glucose transporter (GLUT) 1, 3, and 4 gene expressions in the hippocampus and spatial memory in a streptozotocin (STZ)-induced AD rat model was investigated in the present study. Materials and Methods Fifty-six adult male Sprague-Dawley rats (280±20 g) were allocated into eight distinct groups (n=7) of five controls (negative, Ins, Cinn, Ins+Cinn, and STZs) and three treatments (STZ+ Ins, STZ+ Cinn, and STZ+ Ins + Cinn). Single dose STZ 4 mg/kg (icv), Cinn at a dose of 200 mg/ kg (orally for 14 days), and Ins 5 mIU/5 µl (icv for 14 days) were administered in the defined groups. To evaluate the behavioral performance the animals were subjected to the Morris Water Maze (MWM) test. The level of mRNA expression of GLUTs was evaluated by the Real time-PCR method. Results In the STZ+Cinn+Ins group, the performance of animals in the MWM test was improved and the over-expression of GLUTs genes in hippocampal tissue was observed. The results of Ins and Cinn synergist treatment groups revealed improvement in the behavioral tests and gene expression compared with Ins and Cinn treatment groups (P<0.001). Conclusion Administration of Ins and Cinn has a positive effect on the function of the AD rat model. To clarify the effect of Ins and Cinn extract on the GLUTs investigated in this study, it is essential to evaluate their influence on the protein levels.
Collapse
Affiliation(s)
- Elham Sajadi
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Javad Sajedianfard
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Saeid Hosseinzadeh
- Department of Food Hygiene and Public Health, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Mahnaz Taherianfard
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
31
|
Woodfield A, Gonzales T, Helmerhorst E, Laws S, Newsholme P, Porter T, Verdile G. Current Insights on the Use of Insulin and the Potential Use of Insulin Mimetics in Targeting Insulin Signalling in Alzheimer's Disease. Int J Mol Sci 2022; 23:15811. [PMID: 36555450 PMCID: PMC9779379 DOI: 10.3390/ijms232415811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/10/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease (AD) and type 2 diabetes (T2D) are chronic diseases that share several pathological mechanisms, including insulin resistance and impaired insulin signalling. Their shared features have prompted the evaluation of the drugs used to manage diabetes for the treatment of AD. Insulin delivery itself has been utilized, with promising effects, in improving cognition and reducing AD related neuropathology. The most recent clinical trial involving intranasal insulin reported no slowing of cognitive decline; however, several factors may have impacted the trial outcomes. Long-acting and rapid-acting insulin analogues have also been evaluated within the context of AD with a lack of consistent outcomes. This narrative review provided insight into how targeting insulin signalling in the brain has potential as a therapeutic target for AD and provided a detailed update on the efficacy of insulin, its analogues and the outcomes of human clinical trials. We also discussed the current evidence that warrants the further investigation of the use of the mimetics of insulin for AD. These small molecules may provide a modifiable alternative to insulin, aiding in developing drugs that selectively target insulin signalling in the brain with the aim to attenuate cognitive dysfunction and AD pathologies.
Collapse
Affiliation(s)
- Amy Woodfield
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Australia
| | - Tatiana Gonzales
- Curtin Medical School, Curtin University, Bentley 6102, Australia
| | - Erik Helmerhorst
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Australia
| | - Simon Laws
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup 6027, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup 6027, Australia
| | - Philip Newsholme
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Australia
| | - Tenielle Porter
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup 6027, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup 6027, Australia
| | - Giuseppe Verdile
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup 6027, Australia
| |
Collapse
|
32
|
Hunt NJ, Wahl D, Westwood LJ, Lockwood GP, Le Couteur DG, Cogger VC. Targeting the liver in dementia and cognitive impairment: Dietary macronutrients and diabetic therapeutics. Adv Drug Deliv Rev 2022; 190:114537. [PMID: 36115494 PMCID: PMC10125004 DOI: 10.1016/j.addr.2022.114537] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 01/24/2023]
Abstract
Many people living with dementia and cognitive impairment have dysfunctional mitochondrial and insulin-glucose metabolism resembling type 2 diabetes mellitus and old age. Evidence from human trials shows that nutritional interventions and anti-diabetic medicines that target nutrient-sensing pathways overcome these deficits in glucose and energy metabolism and can improve cognition and/or reduce symptoms of dementia. The liver is the main organ that mediates the systemic effects of diets and many diabetic medicines; therefore, it is an intermediate target for such dementia interventions. A challenge is the efficacy of these treatments in older age. Solutions include the targeted hepatic delivery of diabetic medicines using nanotechnologies and titration of macronutrients to optimize hepatic energy metabolism.
Collapse
Affiliation(s)
- Nicholas J Hunt
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2008, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW 2008, Australia; Sydney Nano Institute, The University of Sydney, Sydney, NSW 2008, Australia; ANZAC Research Institute & Centre for Education and Research on Ageing, Concord Repatriation General Hospital, Concord, NSW 2139, Australia
| | - Devin Wahl
- Department of Health and Exercise Science & Centre for Healthy Aging, Colorado State University, CO 80523, United States
| | - Lara J Westwood
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2008, Australia; ANZAC Research Institute & Centre for Education and Research on Ageing, Concord Repatriation General Hospital, Concord, NSW 2139, Australia
| | - Glen P Lockwood
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2008, Australia; ANZAC Research Institute & Centre for Education and Research on Ageing, Concord Repatriation General Hospital, Concord, NSW 2139, Australia
| | - David G Le Couteur
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2008, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW 2008, Australia; ANZAC Research Institute & Centre for Education and Research on Ageing, Concord Repatriation General Hospital, Concord, NSW 2139, Australia
| | - Victoria C Cogger
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2008, Australia; ANZAC Research Institute & Centre for Education and Research on Ageing, Concord Repatriation General Hospital, Concord, NSW 2139, Australia.
| |
Collapse
|
33
|
Lee K, Chen J, Wang C. Association between diabetes mellitus and post-stroke cognitive impairment. J Diabetes Investig 2022; 14:6-11. [PMID: 36181402 PMCID: PMC9807143 DOI: 10.1111/jdi.13914] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/29/2022] [Accepted: 09/07/2022] [Indexed: 01/07/2023] Open
Abstract
Stroke survivors suffer from various physical, emotional, and cognitive impairments. These changes are dynamic and depend on multiple factors, including underlying diseases, baseline brain function and pathology, the site of the stroke and the post-stroke inflammation, neurogenesis as well as the subsequent remodeling of the neuro-network. First we review the structural and pathological changes of the brain in stroke survivors with diabetes mellitus, which may lead to post-stroke cognitive dysfunction. Second, we provide evidence of hyperglycemia, diabetes mellitus, hypoglycemia, and their relationship with post-stroke cognitive impairment (PSCI) and post-stroke dementia (PSD). In addition to conventional biomarkers, such as HbA1c, we also provide other novel tools to predict PSCI/PSD, such as glycemic variability, receptor for advanced glycation end products, and gut microbiota. Finally, we attempt to provide some modifying methods for glycemic control, focusing on the prevention of PSCI/PSD.
Collapse
Affiliation(s)
- Kang‐Po Lee
- College of MedicineI‐Shou UniversityKaohsiungTaiwan,Stroke Center and Department of NeurologyE‐Da HospitalKaohsiungTaiwan
| | | | - Chih‐Yuan Wang
- Division of Endocrinology and Metabolism, Department of Internal MedicineCollege of Medicine, National Taiwan University Hospital, National Taiwan UniversityTaipeiTaiwan
| |
Collapse
|
34
|
Newby D, Linden AB, Fernandes M, Molero Y, Winchester L, Sproviero W, Ghose U, Li QS, Launer LJ, Duijn CMV, Nevado-Holgado AJ. Comparative effect of metformin versus sulfonylureas with dementia and Parkinson's disease risk in US patients over 50 with type 2 diabetes mellitus. BMJ Open Diabetes Res Care 2022; 10:10/5/e003036. [PMID: 36109050 PMCID: PMC9478804 DOI: 10.1136/bmjdrc-2022-003036] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/28/2022] [Indexed: 11/26/2022] Open
Abstract
INTRODUCTION Type 2 diabetes is a risk factor for dementia and Parkinson's disease (PD). Drug treatments for diabetes, such as metformin, could be used as novel treatments for these neurological conditions. Using electronic health records from the USA (OPTUM EHR) we aimed to assess the association of metformin with all-cause dementia, dementia subtypes and PD compared with sulfonylureas. RESEARCH DESIGN AND METHODS A new user comparator study design was conducted in patients ≥50 years old with diabetes who were new users of metformin or sulfonylureas between 2006 and 2018. Primary outcomes were all-cause dementia and PD. Secondary outcomes were Alzheimer's disease (AD), vascular dementia (VD) and mild cognitive impairment (MCI). Cox proportional hazards models with inverse probability of treatment weighting (IPTW) were used to estimate the HRs. Subanalyses included stratification by age, race, renal function, and glycemic control. RESULTS We identified 96 140 and 16 451 new users of metformin and sulfonylureas, respectively. Mean age was 66.4±8.2 years (48% male, 83% Caucasian). Over the 5-year follow-up, 3207 patients developed all-cause dementia (2256 (2.3%) metformin, 951 (5.8%) sulfonylurea users) and 760 patients developed PD (625 (0.7%) metformin, 135 (0.8%) sulfonylurea users). After IPTW, HRs for all-cause dementia and PD were 0.80 (95% CI 0.73 to 0.88) and 1.00 (95% CI 0.79 to 1.28). HRs for AD, VD and MCI were 0.81 (0.70-0.94), 0.79 (0.63-1.00) and 0.91 (0.79-1.04). Stronger associations were observed in patients who were younger (<75 years old), Caucasian, and with moderate renal function. CONCLUSIONS Metformin users compared with sulfonylurea users were associated with a lower risk of all-cause dementia, AD and VD but not with PD or MCI. Age and renal function modified risk reduction. Our findings support the hypothesis that metformin provides more neuroprotection for dementia than sulfonylureas but not for PD, but further work is required to assess causality.
Collapse
Affiliation(s)
- Danielle Newby
- Psychiatry, University of Oxford, Oxford, UK
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | | | | | - Yasmina Molero
- Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institute, Stockholm, Sweden
- Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | | | | | | | - Qingqin S Li
- Neuroscience, Janssen Research and Development, Titusville, New Jersey, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Science, National Institute on Aging, Bethesda, Maryland, USA
| | - Cornelia M van Duijn
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Department of Epidemiology, Erasmus MC, Rotterdam, Netherlands
| | | |
Collapse
|
35
|
Zhang T, Shaw M, Cherbuin N. Association between Type 2 Diabetes Mellitus and Brain Atrophy: A Meta-Analysis (Diabetes Metab J 2022;46:781-802). Diabetes Metab J 2022; 46:815-816. [PMID: 36193730 PMCID: PMC9532180 DOI: 10.4093/dmj.2022.0296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Tianqi Zhang
- Centre for Research on Ageing, Health and Wellbeing, The Australian National University, Canberra, Australia
| | - Marnie Shaw
- Centre for Research on Ageing, Health and Wellbeing, The Australian National University, Canberra, Australia
| | - Nicolas Cherbuin
- Centre for Research on Ageing, Health and Wellbeing, The Australian National University, Canberra, Australia
- Corresponding author: Nicolas Cherbuin https://orcid.org/0000-0001-6481-0748 Centre for Research on Ageing, Health and Wellbeing, Florey Building, 54 Mills Road, Australian National University, Canberra, ACT 2601, Australia E-mail:
| |
Collapse
|
36
|
Zhang JH, Zhang XY, Sun YQ, Lv RH, Chen M, Li M. Metformin use is associated with a reduced risk of cognitive impairment in adults with diabetes mellitus: A systematic review and meta-analysis. Front Neurosci 2022; 16:984559. [PMID: 36090264 PMCID: PMC9453211 DOI: 10.3389/fnins.2022.984559] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 07/29/2022] [Indexed: 11/23/2022] Open
Abstract
Objective Controversy exists regarding the impact of metformin and whether it prevents or promotes the incidence of cognitive dysfunction. This systematic review and meta-analysis were conducted to identify the effect of metformin therapy on cognitive function in patients with diabetes. Methods Electronic databases (PubMed, EMBASE, PsycINFO, the Cochrane Library, and Web of Science) were systematically searched by two investigators from the date of inception until March 1, 2022. The study followed PRISMA guidelines. Inclusion criteria were defined according to the PECOSmodel. Eligible studies investigated cognitive dysfunction in metformin users compared with non-users in adults with diabetes. Only observational study designs (such as cohort, cross-section, and case-control) were included. Results A systematic search identified 1,839 articles, of which 28 (17 cohort, 8 case-control, and 3 cross-sectional studies) were included in the meta-analysis. Metformin reduced the occurrence of cognitive impairment in patients with diabetes [unadjusted hazard ratio (HR) = 0.67, 95% CI: 0.62–0.73; adjusted hazard ratio (aHR) = 0.92, 95% CI: 0.85–0.99]. In addition, the use of metformin was associated with a decreased risk of dementia (HR = 0.64, 95% CI: 0.59–0.69; aHR = 0.90, 95% CI: 0.84–0.96), while a random-effects meta-analysis indicated no significant effect of metformin on the risk of Alzheimer's disease (AD) (HR = 0.85, 95% CI: 0.60–1.22; aHR = 1.10, 95% CI: 0.95–1.28). Conclusion Metformin therapy decreased the occurrence risk of cognitive decline in patients with diabetes mellitus. Moreover, the use of metformin by adults with diabetes for the prevention of dementia, but not AD, is supported by the available evidence.
Collapse
Affiliation(s)
- Jia-Hao Zhang
- Laboratory of Laser Sports Medicine, School of Sports Science, South China Normal University, Guangzhou, China
| | - Xin-Yang Zhang
- Laboratory of Laser Sports Medicine, School of Sports Science, South China Normal University, Guangzhou, China
- *Correspondence: Xin-Yang Zhang
| | - Yan-Qiu Sun
- Department of Rehabilitation Medicine, Guangdong Women and Children Hospital, Guangzhou, China
| | - Ren-Hua Lv
- Department of Rehabilitation Medicine, Xiangtan Central Hospital, Xiangtan, China
| | - Mei Chen
- Laboratory of Laser Sports Medicine, School of Sports Science, South China Normal University, Guangzhou, China
| | - Meng Li
- Laboratory of Laser Sports Medicine, School of Sports Science, South China Normal University, Guangzhou, China
| |
Collapse
|
37
|
Dai J, Ports KD, Corrada MM, Odegaard AO, O’Connell J, Jiang L. Metformin and Dementia Risk: A Systematic Review with Respect to Time Related Biases. J Alzheimers Dis Rep 2022; 6:443-459. [PMID: 36186728 PMCID: PMC9484147 DOI: 10.3233/adr-220002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND When studying drug effects using observational data, time-related biases may exist and result in spurious associations. Numerous observational studies have investigated metformin and dementia risk, but have reported inconsistent findings, some of which might be caused by unaddressed time-related biases. Immortal time bias biases the results toward a "protective" effect, whereas time-lag and time-window biases can lead to either a "detrimental" or "protective" effect. OBJECTIVE To conduct a systematic review examining time-related biases in the literature on metformin and dementia. METHODS The electronic databases PubMed, Web of Science, and ProQuest were searched for the terms "Metformin" AND ("dementia" OR "Alzheimer's Disease" OR "cognitive impairment"). These databases were searched from inception through 09/24/2021. Only English language articles and human research were eligible. RESULTS Seventeen studies were identified: thirteen cohort studies, two case-control studies, and two nested case-control studies. Eleven (64.7%) studies reported a reduced risk of dementia associated with metformin use; two (11.8%) suggested metformin increased dementia risk, while four (23.5%) concluded no significant associations. Eight (61.5%) of thirteen cohort studies had immortal time bias or did not clearly address it. Fifteen (88.2%) of seventeen reviewed studies had time-lag bias or did not clearly address it. Two (50.0%) of four case-control studies did not explicitly address time-window bias. The studies that addressed most biases concluded no associations between metformin and dementia risk. CONCLUSION None of the reviewed studies clearly addressed relevant time-related biases, illustrating time-related biases are common in observational studies investigating the impact of anti-diabetic medications on dementia risk.
Collapse
Affiliation(s)
- Jiahui Dai
- Department of Epidemiology & Biostatistics, University of California Irvine, Irvine, CA, USA
| | - Kayleen Deanna Ports
- Department of Epidemiology & Biostatistics, University of California Irvine, Irvine, CA, USA
| | - Maria M. Corrada
- Department of Epidemiology & Biostatistics, University of California Irvine, Irvine, CA, USA
- Department of Neurology, University of California Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California Irvine, Irvine, CA, USA
| | - Andrew O. Odegaard
- Department of Epidemiology & Biostatistics, University of California Irvine, Irvine, CA, USA
| | - Joan O’Connell
- Centers for American Indian and Alaska Native Health, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Luohua Jiang
- Department of Epidemiology & Biostatistics, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
38
|
Ji S, Zhao X, Zhu R, Dong Y, Huang L, Zhang T. Metformin and the risk of dementia based on an analysis of 396,332 participants. Ther Adv Chronic Dis 2022; 13:20406223221109454. [PMID: 35847477 PMCID: PMC9277541 DOI: 10.1177/20406223221109454] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 06/01/2022] [Indexed: 11/17/2022] Open
Abstract
Background: AMPK has attracted widespread interest as a potential therapeutic target for age-related diseases, given its key role in controlling energy homeostasis. Metformin (Met) has historically been used to treat Type 2 diabetes and has been shown to counteract age-related diseases. However, studies regarding the relationship between Met and a variety of age-related classifications of cognitive decline have reported mixed findings. Objective: To assess the potential effect of Met on the onset of dementia and discuss the possible biological mechanisms involved. Methods: This study was registered in the PROSPERO database (CRD420201251468). PubMed, Embase, and Cochrane Library were searched from inception to 25 May 2021, for population-based cohort studies. Effect estimates with 95% confidence intervals (CIs) were pooled using the random-effects model. Meta-regression and subgroup analyses were performed to explore sources of heterogeneity and the stability of the results. Results: Fourteen population-based cohort studies (17 individual comparisons) involving 396,332 participants were identified. Meta-analysis showed that Met exposure was significantly associated with reduced risk of all subtypes of dementias [relative risk (RR) = 0.79, 95% CI = 0.68–0.91; p < 0.001]. Conversely, no significant reduction in risk was observed for those who received Met monotherapy at the onset of vascular dementia (VD), Parkinson’s disease (PD), and Alzheimer’s disease (AD). The effect was more prominent in patients who had long-term Met exposure (⩾4 years) (RR = 0.38, 95% CI = 0.32–0.46; p < 0.001), while no such significant effect was found with short-term Met exposure (1–2 years) (RR = 1.20, 95% CI = 0.87–1.66; p < 0.001). Moreover, no association was observed for Met exposure in participants of European descent (RR = 1.01, 95% CI = 0.66–1.54; p = 0.003) compared with those from other countries. Conclusion: Based on the evidence from population-based cohort studies, our findings suggest that the AMPK activator, Met, is a potential geroprotective agent for dementias, particularly among long-term Met users. Due to the significant heterogeneity among the included studies, we should interpret the results with caution.
Collapse
Affiliation(s)
- Shiliang Ji
- Department of pharmacy, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Xingxing Zhao
- Department of Neonatology, The Affiliated Suzhou Hospital of Nanjing Medical University (Suzhou Municipal Hospital), Gusu School, Nanjing Medical University, Suzhou, China
| | - Ruifang Zhu
- Department of pharmacy, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Yongchao Dong
- Department of pharmacy, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Lifeng Huang
- Department of pharmacy, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou 215153, China
| | - Taiquan Zhang
- Department of pharmacy, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou 215153, China
| |
Collapse
|
39
|
Luo A, Ning P, Lu H, Huang H, Shen Q, Zhang D, Xu F, Yang L, Xu Y. Association Between Metformin and Alzheimer's Disease: A Systematic Review and Meta-Analysis of Clinical Observational Studies. J Alzheimers Dis 2022; 88:1311-1323. [PMID: 35786654 DOI: 10.3233/jad-220180] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND As one of the widely used drugs for the management of type 2 diabetes mellites (T2DM), metformin is increasingly believed to delay cognitive deterioration and therapeutically for Alzheimer's disease (AD) patients especially those with T2DM. However, studies of the potential neuroprotective effects of metformin in AD patients have reported contradictory results. OBJECTIVE This study aimed to evaluate the association between metformin and the risk of developing AD. METHODS We systematically searched the PubMed, EMBASE, Web of Science, Cochrane Central Register of Controlled Trials, and ClinicalTrials.gov databases to identify clinical observational studies on the relationship between AD risk and metformin use published before December 20, 2021. Two investigators independently screened records, extracted data, and assessed the quality of the studies. Pooled odds ratios (ORs) and corresponding 95% confidence intervals (CIs) were calculated using random-effect models. RESULTS After screening a total of 1,670 records, we included 10 studies involving 229,110 participants. The meta-analysis showed no significant association between AD incidence and metformin exposure (OR 1.17, 95% CI 0.88-1.56, p = 0.291). However, subgroup analysis showed that among Asians, the risk of AD was significantly higher among metformin users than those who did not (OR 1.71, 95% CI 1.24-2.37, p = 0.001). CONCLUSION The available evidence does not support the idea that metformin reduces risk of AD, and it may, in fact, increase the risk in Asians. Further well-designed randomized controlled trials are required to understand the role played by metformin and other antidiabetic drugs in the prevention of AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Anling Luo
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Pingping Ning
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Haitao Lu
- Department of Neurology, Third People's Hospital of Chengdu, Chengdu, Sichuan Province, P.R. China
| | - Hongyan Huang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Qiuyan Shen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Dan Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Fang Xu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Li Yang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, P.R. China
| | - Yanming Xu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, P.R. China
| |
Collapse
|
40
|
Zhang Y, Zhang Y, Shi X, Han J, Lin B, Peng W, Mei Z, Lin Y. Metformin and the risk of neurodegenerative diseases in patients with diabetes: A meta-analysis of population-based cohort studies. Diabet Med 2022; 39:e14821. [PMID: 35213749 DOI: 10.1111/dme.14821] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 02/23/2022] [Indexed: 11/29/2022]
Abstract
AIMS The association between metformin use and neurodegenerative disease (ND) onset remains controversial. In this systematic review and meta-analysis, we aimed to determine the relationship between metformin use and ND risk based on data from population-based cohort studies. METHODS Articles were systematically searched in PubMed, EMBASE and Cochrane Library databases. Pooled relative risks (RRs) with 95% CIs were obtained using a random-effects model. Subgroup analyses, sensitivity analyses and meta-regression were performed to identify the sources of heterogeneity and strengthen the results. RESULTS Twelve population-based cohort studies involving 194,792 participants (94,462 metformin users and 100,330 metformin non-users) were eligible for inclusion in this meta-analysis. The pooled RR of NDs reached 0.77 (95% CI 0.67-0.88) when comparing metformin users with non-users. The effects were more prominent in long-term metformin users (≥4 years) (RR 0.29, 95% CI 0.13-0.44) and studies from Asian countries (RR 0.69, 95% CI 0.64-0.74). The effect estimates were stable when stratified by subtypes of NDs, study designs, and control definitions (p for interaction >0.05). Meta-regression did not identify the coefficients as the sources of heterogeneity (all p > 0.05). CONCLUSIONS This systematic review and meta-analysis found that metformin use, especially long-term use, was associated with lower ND risk. However, because there was substantial heterogeneity among studies, high-quality randomized controlled trials are still needed to confirm this finding.
Collapse
Affiliation(s)
- Yunnan Zhang
- Department of Pharmacy, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Yi Zhang
- Department of Pharmacy, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Xiujin Shi
- Department of Pharmacy, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jialun Han
- Department of Pharmacy, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Baidi Lin
- Department of Pharmacy, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Wenxing Peng
- Department of Pharmacy, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Zubing Mei
- Department of Anorectal Surgery, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Anorectal Disease Institute of Shuguang Hospital, Shanghai, China
| | - Yang Lin
- Department of Pharmacy, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
41
|
Wu CY, Shapiro L, Ouk M, MacIntosh BJ, Black SE, Shah BR, Swardfager W. Glucose-lowering drugs, cognition, and dementia: The clinical evidence. Neurosci Biobehav Rev 2022; 137:104654. [PMID: 35398114 DOI: 10.1016/j.neubiorev.2022.104654] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/01/2022] [Accepted: 04/03/2022] [Indexed: 11/19/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is an important risk factor for dementia. The possibility to mitigate this risk by controlling T2DM is compelling; however, different glucose-lowering drugs have different effects on the brain by virtue of their different mechanisms of action. The clinical and epidemiological data appear mixed, warranting careful critical evaluation of the human studies. Here we examine the evidence in the context of dementia prevention and treatment, both for people with and without T2DM. We discuss the evidence on this scaffold of research directions, identifying methodological complexities in the extant literature (e.g. comparator discrepancies, changes in the therapeutic landscape), and the implications of different outcome measures (e.g. neuropsychological). We consider possible implications of cerebrovascular protection vs. effects on progression of neurodegenerative proteinopathy, and we present a research roadmap for glucose-lowering drugs in cognitive neurology, including neuroimaging, and fluid biomarkers. We conclude that there is great potential to advance personalized strategies to prevent and treat dementia with glucose-lowering drugs.
Collapse
Affiliation(s)
- Che-Yuan Wu
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Lila Shapiro
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Michael Ouk
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Bradley J MacIntosh
- Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Canadian Partnership for Stroke Recovery, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Sandra E Black
- Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada; Canadian Partnership for Stroke Recovery, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medicine, Division of Neurology, University of Toronto, Toronto, Ontario, Canada; Toronto Dementia Research Alliance, Toronto, Ontario, Canada
| | - Baiju R Shah
- ICES, Toronto, Ontario, Canada; Divisions of Endocrinology and Obstetric Medicine, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Walter Swardfager
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada; Canadian Partnership for Stroke Recovery, Sunnybrook Research Institute, Toronto, Ontario, Canada; KITE UHN Toronto Rehabilitation Institute, Toronto, Ontario, Canada
| |
Collapse
|
42
|
Torrandell-Haro G, Branigan GL, Brinton RD, Rodgers KE. Association Between Specific Type 2 Diabetes Therapies and Risk of Alzheimer’s Disease and Related Dementias in Propensity-Score Matched Type 2 Diabetic Patients. Front Aging Neurosci 2022; 14:878304. [PMID: 35601622 PMCID: PMC9120543 DOI: 10.3389/fnagi.2022.878304] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/15/2022] [Indexed: 11/13/2022] Open
Abstract
Objective We sought to determine the impact of Type 2 Diabetes Mellitus (T2D) anti-hyperglycemic medications (A-HgM) on risk of Alzheimer’s disease (AD) and related dementias (ADRD) outcomes including vascular dementia, and non-AD dementia such as frontotemporal, Lewy body, and mixed etiology dementias. Research Design and Methods This retrospective cohort study used the US-based Mariner claims dataset. 1,815,032 T2D participants 45 years and older with records 6 months prior and at least 3 years after the diagnosis of T2D were included. Claims were surveyed for a diagnosis of AD and ADRD 12 months post T2D diagnosis. A propensity score approach was used to minimize selection bias. Analyses were conducted between January 1st and February 28th, 2021. Results In this cohort study A-HgM exposure was associated with decreased diagnosis of AD (RR, 0.61; 95% CI, 0.59–0.62; p < 0.001), vascular dementia (RR, 0.72; 95% CI, 0.69–0.74; p < 0.001) and non-AD dementia (RR, 0.67; 95% CI, 0.66–0.68; p < 0.001). Metformin was associated with the greatest risk reduction and insulin with the least reduction in risk compared to patients not receiving A-HgM for ADRD risk. Of interest, patients with a diagnosis of AD, while either on metformin or insulin, were older in age and predominately female, than individuals on these drugs that did not develop AD. Mean (SD) follow-up was 6.2 (1.8) years. Conclusion After controlling for age, sex, and comorbidities, A-HgM in patients with T2D was associated with a reduced risk of AD and ADRD. These findings provide evidence in support of T2D as a risk factor for AD and ADRD and the beneficial impact of early and effective control of hyperglycemia to mitigate risk.
Collapse
Affiliation(s)
- Georgina Torrandell-Haro
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, United States
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Gregory L. Branigan
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, United States
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
- MD-PhD Training Program, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Roberta Diaz Brinton
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, United States
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
- Department of Neurology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Kathleen E. Rodgers
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, United States
- Department of Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
- *Correspondence: Kathleen E. Rodgers,
| |
Collapse
|
43
|
Ning P, Luo A, Mu X, Xu Y, Li T. Exploring the dual character of metformin in Alzheimer's disease. Neuropharmacology 2022; 207:108966. [PMID: 35077762 DOI: 10.1016/j.neuropharm.2022.108966] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 12/17/2021] [Accepted: 01/17/2022] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, which results in dementia typically in the elderly. The disease is mainly characterized by the deposition of amyloid beta (Aβ) plaques and neurofibrillary tangles (NFTs) in the brain. However, only few drugs are available for AD because of its unknown pathological mechanism which limits the development of new drugs. Therefore, it is urgent to identify potential therapeutic strategies for AD. Moreover, research have showed that there is a significant association between Type 2 diabetes mellites (T2DM) and AD, suggesting that the two diseases may share common pathophysiological mechanisms. Such mechanisms include impaired insulin signaling, altered glucose metabolism, inflammation, oxidative stress, and premature aging, which strongly affect cognitive function and increased risk of dementia. Consequently, as a widely used drug for T2DM, metformin also has therapeutic potential for AD in vivo. It has been confirmed that metformin is beneficial on the brain of AD animal models. The mechanisms underlying the effects of metformin in Alzheimer's disease are complex and multifaceted. Metformin may work through mechanisms involving homeostasis of glucose metabolism, decrease of amyloid plaque deposition, normalization of tau protein phosphorylation and enhancement of autophagy. However, in clinical trials, metformin had little effects on patients with mild cognitive impairment or mild AD. Pathological effects and negative clinical results of metformin on AD make the current topic quite controversial. By reviewing the latest progress of related research, this paper summarizes the possible role of metformin in AD. The purpose of this study is not only to determine the potential treatment of AD, but also other related neurodegenerative diseases.
Collapse
Affiliation(s)
- Pingping Ning
- Department of Neurology, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan Province, 610041, PR China.
| | - Anling Luo
- Department of Neurology, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan Province, 610041, PR China.
| | - Xin Mu
- Department of Neurology, Chengdu First People's Hospital, 18 Wanxiang North Road, Chengdu, Sichuan Province, 610041, PR China.
| | - Yanming Xu
- Department of Neurology, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan Province, 610041, PR China.
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University/Air Force Medical University, No. 169 Changle West Rd, Xi'an, 710032, PR China.
| |
Collapse
|
44
|
Moran C, Than S, Callisaya M, Beare R, Srikanth V. New Horizons-Cognitive Dysfunction Associated With Type 2 Diabetes. J Clin Endocrinol Metab 2022; 107:929-942. [PMID: 34788847 DOI: 10.1210/clinem/dgab797] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Indexed: 11/19/2022]
Abstract
The prevalence of type 2 diabetes (T2D) and cognitive dysfunction increases with age. As society ages, clinicians will be increasingly tasked with managing older people who have both T2D and cognitive dysfunction. T2D is associated with an increased risk of cognitive dysfunction and hence there is increasing interest in whether T2D is a causal factor in the pathogenesis of cognitive decline and dementia. Recent advances in the use of sensitive measures of in vivo brain dysfunction in life-course studies can help understand potential mechanistic pathways and also help guide recommendations for clinical practice. In this article we will describe new horizons in the understanding of cognitive dysfunction associated with T2D. Coming from a clinical perspective, we discuss potential mechanisms and pathways linking the 2 conditions and the contribution of multimodal neuroimaging and study designs to advancing understanding in the field. We also highlight the important issues on the horizon that will need addressing in clinical identification, management, and risk reduction for people with coexistent T2D and cognitive dysfunction.
Collapse
Affiliation(s)
- Chris Moran
- Academic Unit, Peninsula Clinical School, Central Clinical School, Monash University, Melbourne, 3199 Victoria, Australia
- Department of Geriatric Medicine, Peninsula Health, Melbourne, 3199 Victoria, Australia
- Department of Geriatric Medicine, Alfred Health, Melbourne, 3004 Victoria, Australia
| | - Stephanie Than
- Academic Unit, Peninsula Clinical School, Central Clinical School, Monash University, Melbourne, 3199 Victoria, Australia
- Department of Geriatric Medicine, Peninsula Health, Melbourne, 3199 Victoria, Australia
| | - Michele Callisaya
- Academic Unit, Peninsula Clinical School, Central Clinical School, Monash University, Melbourne, 3199 Victoria, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, 7000 Tasmania, Australia
| | - Richard Beare
- Academic Unit, Peninsula Clinical School, Central Clinical School, Monash University, Melbourne, 3199 Victoria, Australia
- Developmental Imaging, Murdoch Children's Research Institute, Melbourne, 3052 Victoria, Australia
| | - Velandai Srikanth
- Academic Unit, Peninsula Clinical School, Central Clinical School, Monash University, Melbourne, 3199 Victoria, Australia
- Department of Geriatric Medicine, Peninsula Health, Melbourne, 3199 Victoria, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, 7000 Tasmania, Australia
| |
Collapse
|
45
|
Secnik J, Xu H, Schwertner E, Hammar N, Alvarsson M, Winblad B, Eriksdotter M, Garcia-Ptacek S, Religa D. The association of antidiabetic medications and Mini-Mental State Examination scores in patients with diabetes and dementia. Alzheimers Res Ther 2021; 13:197. [PMID: 34857046 PMCID: PMC8641148 DOI: 10.1186/s13195-021-00934-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/09/2021] [Indexed: 02/06/2023]
Abstract
Background The effect of antidiabetic medication on cognitive function is unclear. We analyzed the association between five antidiabetic drugs and change in Mini-Mental State Examination (MMSE) scores in patients with diabetes and dementia. Methods Using the Swedish Dementia Registry and four supplementary Swedish registers/databases, we identified 1873 patients (4732 observations) with diagnosis of type 2 diabetes (diabetes) and Alzheimer’s disease or mixed-pathology dementia who were followed up at least once after dementia diagnosis. Use of metformin, insulin, sulfonylurea, thiazolidinediones (TZD), and dipeptidyl-peptidase-4 inhibitors (DPP-4i) was identified at baseline. Prevalent-user, incident-user, and drug-drug cohorts were sampled, and propensity-score matching was used to analyze comparable subjects. Beta coefficients with 95% confidence intervals (CI) from the random intercept and slope linear mixed-effects models determined the association between the use of antidiabetic medications and decline in MMSE score points between the follow-ups. Inverse-probability weighting was used to account for patient dropout. Results Compared to non-users, prevalent users of metformin (beta 0.89, 95% CI 0.44; 1.33) and DPP-4i (0.72, 0.06; 1.37) experienced a slower cognitive decline with time. Secondly, compared to DPP-4i, the use of insulin (−1.00, −1.95; −0.04) and sulfonylureas (−1.19; −2.33; −0.04) was associated with larger point-wise decrements in MMSE with annual intervals. Conclusions In this large cohort of patients with diabetes and dementia, the use of metformin and DPP-4i was associated with a slower decline in MMSE scores. Further examination of the cognitive effects of metformin and incretin-based medications is warranted. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00934-0.
Collapse
Affiliation(s)
- Juraj Secnik
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Neo, Blickagången 16, 14152, Huddinge, Sweden. .,Department of Neurology, Second Faculty of Medicine, Charles University, Motol University Hospital, Prague, Czech Republic.
| | - Hong Xu
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Neo, Blickagången 16, 14152, Huddinge, Sweden.,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Emilia Schwertner
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Neo, Blickagången 16, 14152, Huddinge, Sweden
| | - Niklas Hammar
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Michael Alvarsson
- Growth and Metabolism, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Winblad
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Maria Eriksdotter
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Neo, Blickagången 16, 14152, Huddinge, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Sara Garcia-Ptacek
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Neo, Blickagången 16, 14152, Huddinge, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Dorota Religa
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Neo, Blickagången 16, 14152, Huddinge, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
46
|
Siervo M, Shannon OM, Llewellyn DJ, Stephan BC, Fontana L. Mediterranean diet and cognitive function: From methodology to mechanisms of action. Free Radic Biol Med 2021; 176:105-117. [PMID: 34562607 DOI: 10.1016/j.freeradbiomed.2021.09.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 12/12/2022]
Abstract
The traditional Mediterranean diet (MedDiet), rich in minimally processed plant foods and fish, has been widely recognized to be one of the healthiest diets. Data from multiple randomized clinical trials have demonstrated its powerful effect against oxidative stress, inflammation and the development and progression of cardiovascular disease, type 2 diabetes, and other metabolic conditions that play a crucial role in the pathogenesis of neurodegenerative diseases. The protecting effects of the MedDiet against cognitive decline have been investigated in several observational and experimental studies. Data from observational studies suggest that the MedDiet may represent an effective dietary strategy for the early prevention of dementia, although these findings require further substantiation in clinical trials which have so far produced inconclusive results. Moreover, as we discuss in this review, accumulating data emphasizes the importance of: 1) maintaining an optimal nutritional and metabolic status for the promotion of healthy cognitive aging, and 2) implementing cognition-sparing dietary and lifestyle interventions during early time-sensitive windows before the pathological cascades turn into an irreversible state. In summary, components of the MedDiet pattern, such as essential fatty acids, polyphenols and vitamins, have been associated with reduced oxidative stress and the current evidence from observational studies seems to assign to the MedDiet a beneficial role in promoting brain health; however, results from clinical trials have been inconsistent. While we advocate for longitudinal analyses and for larger and longer clinical trials to be conducted, we assert our interim support to the use of the MedDiet as a protective dietary intervention for cognitive function based on its proven cardiovascular and metabolic benefits.
Collapse
Affiliation(s)
- Mario Siervo
- School of Life Sciences, The University of Nottingham Medical School, Queen's Medical Centre, Nottingham, UK.
| | - Oliver M Shannon
- Human Nutrition Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - David J Llewellyn
- University of Exeter Medical School, Exeter, UK; Alan Turing Institute, London, UK
| | - Blossom Cm Stephan
- Institute of Mental Health, The University of Nottingham Medical School, Nottingham, UK
| | - Luigi Fontana
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Department of Clinical and Experimental Sciences, Brescia University School of Medicine, Brescia, Italy
| |
Collapse
|
47
|
Kim J, Park JH, Shah K, Mitchell SJ, Cho K, Hoe HS. The Anti-diabetic Drug Gliquidone Modulates Lipopolysaccharide-Mediated Microglial Neuroinflammatory Responses by Inhibiting the NLRP3 Inflammasome. Front Aging Neurosci 2021; 13:754123. [PMID: 34776934 PMCID: PMC8587901 DOI: 10.3389/fnagi.2021.754123] [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: 08/06/2021] [Accepted: 10/07/2021] [Indexed: 12/22/2022] Open
Abstract
The sulfonylurea drug gliquidone is FDA approved for the treatment of type 2 diabetes. Binding of gliquidone to ATP-sensitive potassium channels (SUR1, Kir6 subunit) in pancreatic β-cells increases insulin release to regulate blood glucose levels. Diabetes has been associated with increased levels of neuroinflammation, and therefore the potential effects of gliquidone on micro- and astroglial neuroinflammatory responses in the brain are of interest. Here, we found that gliquidone suppressed LPS-mediated microgliosis, microglial hypertrophy, and proinflammatory cytokine COX-2 and IL-6 levels in wild-type mice, with smaller effects on astrogliosis. Importantly, gliquidone downregulated the LPS-induced microglial NLRP3 inflammasome and peripheral inflammation in wild-type mice. An investigation of the molecular mechanism of the effects of gliquidone on LPS-stimulated proinflammatory responses showed that in BV2 microglial cells, gliquidone significantly decreased LPS-induced proinflammatory cytokine levels and inhibited ERK/STAT3/NF-κB phosphorylation by altering NLRP3 inflammasome activation. In primary astrocytes, gliquidone selectively affected LPS-mediated proinflammatory cytokine expression and decreased STAT3/NF-κB signaling in an NLRP3-independent manner. These results indicate that gliquidone differentially modulates LPS-induced microglial and astroglial neuroinflammation in BV2 microglial cells, primary astrocytes, and a model of neuroinflammatory disease.
Collapse
Affiliation(s)
- Jieun Kim
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu, South Korea
| | - Jin-Hee Park
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu, South Korea
| | - Keshvi Shah
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu, South Korea.,UK-Dementia Research Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Scott John Mitchell
- UK-Dementia Research Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Kwangwook Cho
- UK-Dementia Research Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Hyang-Sook Hoe
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu, South Korea.,Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, South Korea
| |
Collapse
|
48
|
Trout AL, McLouth CJ, Kitzman P, Dobbs MR, Bellamy L, Elkins K, Fraser JF. Hemorrhagic stroke outcomes of KApSR patients with co-morbid diabetes and Alzheimer's disease. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1371. [PMID: 34733923 PMCID: PMC8506530 DOI: 10.21037/atm-21-1451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/07/2021] [Indexed: 11/06/2022]
Abstract
Background Vascular risk factors, such as diabetes mellitus (DM), are associated with poorer outcomes following many neurodegenerative diseases, including hemorrhagic stroke and Alzheimer's disease (AD). Combined AD and DM co-morbidities are associated with an increased risk of hemorrhagic stroke and increased Medicare costs. Therefore, we hypothesized that patients with DM in combination with AD, termed DM/AD, would have increased hemorrhagic stroke severity. Methods Kentucky Appalachian Stroke Registry (KApSR) is a database of demographic and clinical data from patients that live in Appalachia, a distinct region with increased health disparities and stroke severity. Inpatients with a primary indication of hemorrhagic stroke were selected from KApSR for retrospective analysis and were separated into four groups: DM only, AD only, neither, or both. Results Hemorrhagic stroke patients (2,071 total) presented with either intracerebral hemorrhage (ICH), n=1,448, or subarachnoid hemorrhage (SAH), n=623. When examining all four groups, subjects with AD were significantly older (AD+, 80.9±6.6 yrs) (DM+/AD+, 77.4±10.0 yrs) than non AD subjects (DM-/AD-, 61.3±16.5 yrs) and (DM+, 66.0±12.5 yrs). A higher percentage of females were among the AD+ group and a higher percentage of males among the DM+/AD+ group. Interestingly, after adjusting for multiple comparison, DM+/AD+ subjects were ten times as likely to suffer a moderate to severe stroke based on a National Institute of Health Stroke (NIHSS) upon admission [odds ratio (95% CI)] compared to DM-/AD- [0.1 (0.02-0.55)], DM+ [0.11 (0.02-0.59)], and AD+ [0.09(0.01-0.63)]. The odds of DM+/AD+ subjects having an unfavorable discharge destination (death, hospice, long-term care) was significant (P<0.05) from DM-/AD- [0.26 (0.07-0.96)] when adjusting for sex, age, and comorbidities. Conclusions In our retrospective analysis utilizing KApSR, regardless of adjusting for age, sex, and comorbidities, DM+/AD+ patients were significantly more likely to have had a moderate or severe stroke leading to an unfavorable outcome following hemorrhagic stroke.
Collapse
Affiliation(s)
- Amanda L Trout
- Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, USA.,Department of Neurology, University of Kentucky, Lexington, KY, USA
| | | | - Patrick Kitzman
- Department of Behavioral Science, University of Kentucky, Lexington, KY, USA.,HealthCare Stroke Network, Norton Healthcare/UK, Lexington, KY, USA
| | - Michael R Dobbs
- Department of Neurology, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Lisa Bellamy
- HealthCare Stroke Network, Norton Healthcare/UK, Lexington, KY, USA
| | - Kelley Elkins
- HealthCare Stroke Network, Norton Healthcare/UK, Lexington, KY, USA
| | - Justin F Fraser
- Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, USA.,Department of Neurology, University of Kentucky, Lexington, KY, USA.,Department of Neuroscience, University of Kentucky, Lexington, KY, USA.,Department of Neurosurgery, University of Kentucky, Lexington, KY, USA.,Department of Radiology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
49
|
Ribeiro R, Santos AC, Calazans MO, De Oliveira ACP, Vieira LB. Is resveratrol a prospective therapeutic strategy in the co-association of glucose metabolism disorders and neurodegenerative diseases? Nutr Neurosci 2021; 25:2442-2457. [PMID: 34514962 DOI: 10.1080/1028415x.2021.1972514] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Objectives: The mechanism behind the progression of Mild Cognitive Impairment (MCI) to Alzheimer's disease (AD) remains poorly understood. However some evidence pointed out that the co-occurrence of metabolic conditions affecting glucose homeostasis, as type 2 diabetes mellitus (T2DM), may be an important catalyst in this context. Notably, candidate drugs which modulate common pathways in the development of MCI-to-AD mediated by T2DM may offer likely therapy for AD. Nonetheless, limited pharmacological alternatives that modulate common pathways in T2DM, MCI, and AD are available. In the recent decades, studies have shown that resveratrol may act as a neuroprotective compound, but little is known about its potential in improving cognitive and metabolic aspects associated with AD progression mediated by the co-association between TDM2-MCI.Methods: In this review, we discuss possible protective mechanisms of resveratrol on shared pathways associated with AD progression mediated by T2DM-MCI co-occurrence.Results: Some studies indicated that insulin resistance and hyperglycemia may be also a T2DM risk factor for the progression of MCI-to-AD, promoting alterations in metabolic pathways associated with neuronal plasticity, and increasing pro-inflammatory environment. Interestingly, basic research and clinical trials indicate that resveratrol may modulate those pathways, showing a potential neuroprotective effect of this polyphenol.Conclusion: Therefore, there is not enough clinical data supporting the translational therapeutic use of resveratrol in this scenario.
Collapse
Affiliation(s)
- R Ribeiro
- Departamento de Farmacologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - A C Santos
- Departamento de Farmacologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - M O Calazans
- Departamento de Farmacologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - A C P De Oliveira
- Departamento de Farmacologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - L B Vieira
- Departamento de Farmacologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
50
|
Rojas M, Chávez-Castillo M, Bautista J, Ortega Á, Nava M, Salazar J, Díaz-Camargo E, Medina O, Rojas-Quintero J, Bermúdez V. Alzheimer’s disease and type 2 diabetes mellitus: Pathophysiologic and pharmacotherapeutics links. World J Diabetes 2021; 12:745-766. [PMID: 34168725 PMCID: PMC8192246 DOI: 10.4239/wjd.v12.i6.745] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/20/2021] [Accepted: 05/21/2021] [Indexed: 02/06/2023] Open
Abstract
At present, Alzheimer’s disease (AD) and type 2 diabetes mellitus (T2DM) are two highly prevalent disorders worldwide, especially among elderly individuals. T2DM appears to be associated with cognitive dysfunction, with a higher risk of developing neurocognitive disorders, including AD. These diseases have been observed to share various pathophysiological mechanisms, including alterations in insulin signaling, defects in glucose transporters (GLUTs), and mitochondrial dysfunctions in the brain. Therefore, the aim of this review is to summarize the current knowledge regarding the molecular mechanisms implicated in the association of these pathologies as well as recent therapeutic alternatives. In this context, the hyperphosphorylation of tau and the formation of neurofibrillary tangles have been associated with the dysfunction of the phosphatidylinositol 3-kinase and mitogen-activated protein kinase pathways in the nervous tissues as well as the decrease in the expression of GLUT-1 and GLUT-3 in the different areas of the brain, increase in reactive oxygen species, and production of mitochondrial alterations that occur in T2DM. These findings have contributed to the implementation of overlapping pharmacological interventions based on the use of insulin and antidiabetic drugs, or, more recently, azeliragon, amylin, among others, which have shown possible beneficial effects in diabetic patients diagnosed with AD.
Collapse
Affiliation(s)
- Milagros Rojas
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Mervin Chávez-Castillo
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Jordan Bautista
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Ángel Ortega
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Manuel Nava
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Juan Salazar
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Edgar Díaz-Camargo
- Universidad Simón Bolívar, Facultad de Ciencias Jurídicas y Sociales, Cúcuta 540006, Colombia
| | - Oscar Medina
- Universidad Simón Bolívar, Facultad de Ciencias Jurídicas y Sociales, Cúcuta 540006, Colombia
| | - Joselyn Rojas-Quintero
- Pulmonary and Critical Care Medicine Department, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02155, United States
| | - Valmore Bermúdez
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Barranquilla 080001, Colombia
| |
Collapse
|