1
|
Thanh Phuc P, Nguyen PA, Nguyen NN, Hsu MH, Le KN, Tran QV, Huang CW, Yang HC, Chen CY, Le TAH, Le MK, Nguyen HB, Lu CY, Hsu JC. Early detection of Dementia in Type 2 Diabetes population: Predictive analytics using Machine learning approach. J Med Internet Res 2024. [PMID: 39434474 DOI: 10.2196/52107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND The possible association between diabetes mellitus and dementia has raised concerns, given the observed coincidental occurrences. OBJECTIVE This study aims to develop a personalized predictive model, utilizing artificial intelligence, to assess the 5-year and 10-year dementia risk among patients with Type 2 Diabetes Mellitus (T2DM) who are prescribed antidiabetic medications. METHODS This retrospective multicenter study used data from Taipei Medical University Clinical Research Database, which comprises electronic medical records from three hospitals in Taiwan. This study applied eight machine learning algorithms to develop prediction models, including logistic regression (LR), linear discriminant analysis (LDA), gradient boosting machine (GBM), lightGBM (LBGM), AdaBoost, random forest, extreme gradient boosting (XGBoost), and artificial neural network (ANN). These models incorporated a range of variables, encompassing patient characteristics, comorbidities, medication usage, laboratory results, and examination data. RESULTS This study involved a cohort of 43,068 patients diagnosed with T2DM, which accounted for a total of 1,937,692 visits. For model development and validation, 1,300,829 visits were utilized, while an additional 636,863 visits were reserved for external testing. The area under the curve (AUC) of the prediction models range from 0.67 for the logistic regression to 0.98 for the artificial neural networks. Based on the external test results, the model built using the ANN algorithm has the best AUC: 0.97 (5-year follow-up period) and 0.98 (10-year follow-up period). Based on the best model (ANN), age, gender, triglyceride, HbA1c, anti-diabetic agents, stroke history, and other long-term medications were the most important predictors. CONCLUSIONS We have successfully developed a novel computer-aided dementia risk prediction model that can facilitate the clinical diagnosis and management of patients prescribed with antidiabetic medications. However, further investigation is required to assess the model's feasibility and external validity. CLINICALTRIAL
Collapse
Affiliation(s)
- Phan Thanh Phuc
- Taipei Medical University, 250 Wuxing streetXinyi district, Taipei, TW
- University Medical Center Ho Chi Minh City, 215 Hong Bang street, ward 11, district 5, Ho Chi Minh City, VN
| | - Phung-Anh Nguyen
- Taipei Medical University, 250 Wuxing streetXinyi district, Taipei, TW
| | - Nam N Nguyen
- Taipei Medical University, 250 Wuxing streetXinyi district, Taipei, TW
| | - Min-Huei Hsu
- Taipei Medical University, 250 Wuxing streetXinyi district, Taipei, TW
| | - Khanh Nq Le
- Taipei Medical University, 250 Wuxing streetXinyi district, Taipei, TW
| | - Quoc-Viet Tran
- National Taiwan University of Science and Technology, Taipei, TW
| | - Chih-Wei Huang
- Taipei Medical University, 250 Wuxing streetXinyi district, Taipei, TW
| | - Hsuan-Chia Yang
- Taipei Medical University, 250 Wuxing streetXinyi district, Taipei, TW
| | - Cheng-Yu Chen
- Taipei Medical University, 250 Wuxing streetXinyi district, Taipei, TW
| | - Thi Anh Hoa Le
- University Medical Center Ho Chi Minh City, 215 Hong Bang street, ward 11, district 5, Ho Chi Minh City, VN
| | - Minh Khoi Le
- University Medical Center Ho Chi Minh City, 215 Hong Bang street, ward 11, district 5, Ho Chi Minh City, VN
| | - Hoang Bac Nguyen
- University Medical Center Ho Chi Minh City, 215 Hong Bang street, ward 11, district 5, Ho Chi Minh City, VN
| | - Christine Y Lu
- Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, US
- The University of Sydney, Sydney, AU
| | - Jason C Hsu
- Taipei Medical University, 11F, Biomedical Technology Building, No. 301, Yuantong Rd., Zhonghe Dist., New Taipei, TW
| |
Collapse
|
2
|
Zhang J, Tam WWS, Hounsri K, Kusuyama J, Wu VX. Effectiveness of Combined Aerobic and Resistance Exercise on Cognition, Metabolic Health, Physical Function, and Health-related Quality of Life in Middle-aged and Older Adults With Type 2 Diabetes Mellitus: A Systematic Review and Meta-analysis. Arch Phys Med Rehabil 2024; 105:1585-1599. [PMID: 37875170 DOI: 10.1016/j.apmr.2023.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 09/02/2023] [Accepted: 10/09/2023] [Indexed: 10/26/2023]
Abstract
OBJECTIVES To evaluate the effectiveness of combined aerobic and resistance exercise on cognition, metabolic health, physical function, and health-related quality of life (HRQoL) in middle-aged and older adults with type 2 diabetes mellitus (T2DM). DATA SOURCE AND STUDY SELECTION Systematic search of CINAHL, Cochrane, EMBASE, Scopus, PubMed, ProQuest Dissertation and Thesis, PsycINFO, Web of Science databases, and gray literature from Google Scholar. Pertinent randomized controlled trials (RCTs) were selected. The Protocol was registered in the International Prospective Register of Systematic Reviews (PROSPERO CRD42023387336). DATA EXTRACTION The risk of bias was evaluated using the Cochrane Risk of Bias tool by 2 reviewers independently. Outcome data were extracted in a fixed-effect model if heterogeneity test were not significant and I2≤50%; otherwise, the random-effects model was used. DATA SYNTHESIS Sixteen studies with 2426 participants were included in this review. Combined aerobic and resistance exercise had significant positive effects on cognition (SMD=0.34, 95% CI: 0.13 to 0.55), metabolic health on HbA1c (SMD=-0.35, 95% CI: -0.48 to -0.22) and lipid profile (total cholesterol SMD=-0.20, 95% CI: -0.34 to -0.07; low-density lipoprotein SMD=-0.19, 95% CI: -0.33 to -0.05; high-density lipoprotein SMD=0.25, 95% CI: 0.12 to 0.39; and triglycerides SMD=-0.18, 95% CI: -0.31 to -0.04), physical function on aerobic oxygen uptake (SMD=0.58, 95% CI: 0.21 to 0.95) and body mass index (MD=-1.33, 95% CI: -1.84 to -0.82), and physical HRQoL (MD=4.17, 95% CI: 0.86 to 7.48). Our results showed that clinically important effects on cognition may occur in combining the low-moderate intensity of aerobic exercise and progressive intensity of resistance training, the total duration of the exercise needs to be at least 135 minutes per week, among which, resistance training should be at least 60 minutes. CONCLUSION Combined aerobic and resistance exercise effectively improves cognition, ameliorates metabolic health, enhances physical function, and increases physical HRQoL in middle-aged and older adults with T2DM. More RCTs and longitudinal follow-ups are required to provide future evidence of structured combined aerobic and resistance exercise on other domains of cognition.
Collapse
Affiliation(s)
- Jinghua Zhang
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Clinical Research Centre, Singapore
| | - Wilson Wai San Tam
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Clinical Research Centre, Singapore
| | - Kanokwan Hounsri
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Clinical Research Centre, Singapore
| | - Joji Kusuyama
- Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, Tokyo, Japan
| | - Vivien Xi Wu
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Clinical Research Centre, Singapore; NUSMED Healthy Longevity Translational Research Programme, National University of Singapore, Singapore.
| |
Collapse
|
3
|
Bhole RP, Chikhale RV, Rathi KM. Current biomarkers and treatment strategies in Alzheimer disease: An overview and future perspectives. IBRO Neurosci Rep 2024; 16:8-42. [PMID: 38169888 PMCID: PMC10758887 DOI: 10.1016/j.ibneur.2023.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 01/05/2024] Open
Abstract
Alzheimer's disease (AD), a progressive degenerative disorder first identified by Alois Alzheimer in 1907, poses a significant public health challenge. Despite its prevalence and impact, there is currently no definitive ante mortem diagnosis for AD pathogenesis. By 2050, the United States may face a staggering 13.8 million AD patients. This review provides a concise summary of current AD biomarkers, available treatments, and potential future therapeutic approaches. The review begins by outlining existing drug targets and mechanisms in AD, along with a discussion of current treatment options. We explore various approaches targeting Amyloid β (Aβ), Tau Protein aggregation, Tau Kinases, Glycogen Synthase kinase-3β, CDK-5 inhibitors, Heat Shock Proteins (HSP), oxidative stress, inflammation, metals, Apolipoprotein E (ApoE) modulators, and Notch signaling. Additionally, we examine the historical use of Estradiol (E2) as an AD therapy, as well as the outcomes of Randomized Controlled Trials (RCTs) that evaluated antioxidants (e.g., vitamin E) and omega-3 polyunsaturated fatty acids as alternative treatment options. Notably, positive effects of docosahexaenoic acid nutriment in older adults with cognitive impairment or AD are highlighted. Furthermore, this review offers insights into ongoing clinical trials and potential therapies, shedding light on the dynamic research landscape in AD treatment.
Collapse
Affiliation(s)
- Ritesh P. Bhole
- Department of Pharmaceutical Chemistry, Dr. D. Y. Patil institute of Pharmaceutical Sciences & Research, Pimpri, Pune, India
- Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune 411018, India
| | | | - Karishma M. Rathi
- Department of Pharmacy Practice, Dr. D. Y. Patil institute of Pharmaceutical Sciences & Research, Pimpri, Pune, India
| |
Collapse
|
4
|
Zhu W, Tang H, Zhang H, Rajamohan HR, Huang SL, Ma X, Chaudhari A, Madaan D, Almahmoud E, Chopra S, Dodson JA, Brody AA, Masurkar AV, Razavian N. Predicting Risk of Alzheimer's Diseases and Related Dementias with AI Foundation Model on Electronic Health Records. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.26.24306180. [PMID: 38712223 PMCID: PMC11071573 DOI: 10.1101/2024.04.26.24306180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Early identification of Alzheimer's disease (AD) and AD-related dementias (ADRD) has high clinical significance, both because of the potential to slow decline through initiating FDA-approved therapies and managing modifiable risk factors, and to help persons living with dementia and their families to plan before cognitive loss makes doing so challenging. However, substantial racial and ethnic disparities in early diagnosis currently lead to additional inequities in care, urging accurate and inclusive risk assessment programs. In this study, we trained an artificial intelligence foundation model to represent the electronic health records (EHR) data with a vast cohort of 1.2 million patients within a large health system. Building upon this foundation EHR model, we developed a predictive Transformer model, named TRADE, capable of identifying risks for AD/ADRD and mild cognitive impairment (MCI), by analyzing the past sequential visit records. Amongst individuals 65 and older, our model was able to generate risk predictions for various future timeframes. On the held-out validation set, our model achieved an area under the receiver operating characteristic (AUROC) of 0.772 (95% CI: 0.770, 0.773) for identifying the AD/ADRD/MCI risks in 1 year, and AUROC of 0.735 (95% CI: 0.734, 0.736) in 5 years. The positive predictive values (PPV) in 5 years among individuals with top 1% and 5% highest estimated risks were 39.2% and 27.8%, respectively. These results demonstrate significant improvements upon the current EHR-based AD/ADRD/MCI risk assessment models, paving the way for better prognosis and management of AD/ADRD/MCI at scale.
Collapse
Affiliation(s)
- Weicheng Zhu
- NYU, Center for Data Science, New York, NY, 10001, USA
| | - Huanze Tang
- NYU, Center for Data Science, New York, NY, 10001, USA
| | - Hao Zhang
- NYU Grossman School of Medicine, Department of Population Health, New York, NY, 10016, USA
| | | | | | - Xinyue Ma
- NYU, Center for Data Science, New York, NY, 10001, USA
| | | | - Divyam Madaan
- NYU, Courant Institute of Mathematical Sciences, New York, NY, 10001, USA
| | - Elaf Almahmoud
- NYU, Courant Institute of Mathematical Sciences, New York, NY, 10001, USA
| | - Sumit Chopra
- NYU, Courant Institute of Mathematical Sciences, New York, NY, 10001, USA
- NYU Grossman School of Medicine, Department of Radiology, New York, NY, 10016, USA
| | - John A. Dodson
- NYU Grossman School of Medicine, Department of Population Health, New York, NY, 10016, USA
- NYU Grossman School of Medicine, Department of Medicine, New York, NY, 10016, USA
| | - Abraham A. Brody
- NYU Grossman School of Medicine, Department of Medicine, New York, NY, 10016, USA
- NYU Grossman School of Medicine, Rory Meyers College of Nursing, Hartford Institute for Geriatric Nursing, New York, NY, 10016, USA
| | - Arjun V. Masurkar
- NYU Grossman School of Medicine, Department of Neurology, New York, NY, 10016, USA
- NYU Grossman School of Medicine, Department of Neuroscience and Physiology, New York, NY, 10016, USA
- NYU Grossman School of Medicine, Neuroscience Institute, New York, NY, 10016, USA
| | - Narges Razavian
- NYU Grossman School of Medicine, Department of Population Health, New York, NY, 10016, USA
- NYU Grossman School of Medicine, Department of Radiology, New York, NY, 10016, USA
| |
Collapse
|
5
|
Tian R, Liu X, Xiao Y, Jing L, Tao H, Yang L, Meng X. Huang-Lian-Jie-Du decoction drug-containing serum inhibits IL-1β secretion from D-glucose and PA induced BV2 cells via autophagy/NLRP3 signaling. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117686. [PMID: 38160864 DOI: 10.1016/j.jep.2023.117686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/16/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huang-Lian-Jie-Du decoction (HLJDD), a famous traditional Chinese medicine prescription with heat-clearing and detoxifying effects, has been widely used to treat diabetes, dementia, stroke, and other diseases. However, the detailed mechanisms of HLJDD against type 2 diabetes associated cognitive dysfunction (DACD) through inhibiting interleukin-1β (IL-1β) mediated neuroinflammation remain to be further elucidated. AIM OF THE STUDY The aim of this study was to investigate the effect and potential mechanism of HLJDD on IL-1β secretion in a DACD model of BV2 cells induced by D-glucose and palmitic acid (PA). MATERIALS AND METHOD sUltra-performance liquid chromatography-quadrupole/electrostatic field orbital well high-resolution mass spectrometry technology was used to analyze the compounds in HLJDD drug-containing serum. The cytotoxicity was detected by cell counting kit-8. Enzyme-linked immunosorbent assay was used to measure the secretion of IL-1β in BV2 cells. Reactive oxygen species, glutathione, superoxide dismutase, and malondialdehyde kits were used to detect the intracellular oxidative stress levels. The autophagy level was determined by autophagy staining kit and transmission electron microscope. The expression levels of autophagy-related 7 (Atg7), P62, LC3, nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3(NLRP3), Caspase1, and IL-1β were detected by real-time PCR, immunofluorescence, and western blotting. The Atg7siRNA was transfected into BV2 cells to produce autophagy inhibitory effect. Then the effect of HLJDD drug-containing serum on IL-1β secretion in D-glucose and PA induced BV2 cells and the potential mechanism of autophagy-NLRP3 inflammasome activation were further observed. RESULTS Eighty-eight compounds were preliminarily identified in HLJDD drug-containing serum, among which geniposide, baicalin, palmatine, berberine, wogonoside, wogonin, and geniposidic acid were identified as the main prototype components of HLJDD into the blood. In this study, the DACD model of BV2 cells induced by high concentrations of glucose and PA was successfully constructed. HLJDD drug-containing serum significantly reduced the secretion of IL-1β and the activity of NLRP3 inflammasome with improving the oxidative stress level. Interestingly, the enhanced autophagy level was also found. After transfection of Atg7siRNA into BV2 cells, the effect of HLJDD drug-containing serum on autophagy promotion was reversed, but the inhibitory effects on IL-1β secretion, NLRP3 inflammasome activation, and oxidative stress were reduced. CONCLUSIONS These results indicated that the inhibition of HLJDD drug-containing serum on the IL-1β secretion in D-glucose and PA induced BV2 cells was related to autophagy promotion, the decreased NLRP3 inflammasome activation, and the improved oxidative stress. Moreover, the improvement of HLJDD drug-containing serum on IL-1β secretion, NLRP3 inflammasome activation, and oxidative stress were all closely associated with Atg7 mediated autophagy promotion. Geniposide, baicalin, palmatine, berberine, wogonoside, wogonin, and geniposidic acid may be the potential active ingredients of HLJDD drug-containing serum.
Collapse
Affiliation(s)
- Ruimin Tian
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Department of Pharmacology, North Sichuan Medical College, Nanchong, 637000, China
| | - Xianfeng Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yang Xiao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Lijia Jing
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Honglin Tao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Lu Yang
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
6
|
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
|
7
|
Grasset L, Frison E, Helmer C, Catheline G, Chêne G, Dufouil C. Understanding the relationship between type-2 diabetes, MRI markers of neurodegeneration and small vessel disease, and dementia risk: a mediation analysis. Eur J Epidemiol 2024; 39:409-417. [PMID: 38190014 PMCID: PMC11101545 DOI: 10.1007/s10654-023-01080-7] [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: 04/06/2023] [Accepted: 11/03/2023] [Indexed: 01/09/2024]
Abstract
To explore to which extent neurodegeneration and cerebral small vessel disease (SVD) could mediate the association between type-2 diabetes and higher dementia risk. The analytical sample consisted in 2228 participants, out of the Three-City study, aged 65 and older, free of dementia at baseline who underwent brain MRI. Diabetes was defined by medication intake or fasting or non-fasting elevated glucose levels. Dementia status was assessed every 2 to 3 years, during up to 12 years of follow-up. Brain parenchymal fraction (BPF) and white matter hyperintensities volume (WMHV) were selected as markers of neurodegeneration and cerebral SVD respectively. We performed a mediation analysis of the effect of baseline BPF and WMHV (mediators) on the association between diabetes and dementia risk using linear and Cox models adjusted for age, sex, education level, hypertension, hypercholesterolemia, BMI, smoking and alcohol drinking status, APOE-ε4 status, and study site. At baseline, 8.8% of the participants had diabetes. Diabetes (yes vs. no) was associated with higher WMHV (βdiab = 0.193, 95% CI 0.040; 0.346) and lower BPF (βdiab = -0.342, 95% CI -0.474; -0.210), as well as with an increased risk of dementia over 12 years of follow-up (HRdiab = 1.65, 95% CI 1.04; 2.60). The association between diabetes status and dementia risk was statistically mediated by higher WMHV (HRdiab=1.05, 95% CI 1.01; 1.11, mediated part = 10.8%) and lower BPF (HRdiab = 1.12, 95% CI 1.05; 1.20, mediated part = 22.9%). This study showed that both neurodegeneration and cerebral SVD statistically explained almost 30% of the association between diabetes and dementia.
Collapse
Affiliation(s)
- Leslie Grasset
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR 1219, CIC1401-EC, F-33000, Bordeaux, France.
- INSERM U1219, University of Bordeaux, 146 rue Léo Saignat, 33077, Bordeaux cedex, France.
| | - Eric Frison
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR 1219, CIC1401-EC, F-33000, Bordeaux, France
- Service d'Information Médicale, CHU Bordeaux, Bordeaux, France
| | - Catherine Helmer
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR 1219, CIC1401-EC, F-33000, Bordeaux, France
| | - Gwénaëlle Catheline
- INCIA, EPHE, CNRS, Université PSL, University of Bordeaux, 33076, Bordeaux, France
| | - Geneviève Chêne
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR 1219, CIC1401-EC, F-33000, Bordeaux, France
- Pole de sante publique Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000, Bordeaux, France
| | - Carole Dufouil
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR 1219, CIC1401-EC, F-33000, Bordeaux, France
- Pole de sante publique Centre Hospitalier Universitaire (CHU) de Bordeaux, 33000, Bordeaux, France
| |
Collapse
|
8
|
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
|
9
|
Kuate Defo A, Bakula V, Pisaturo A, Labos C, Wing SS, Daskalopoulou SS. Diabetes, antidiabetic medications and risk of dementia: A systematic umbrella review and meta-analysis. Diabetes Obes Metab 2024; 26:441-462. [PMID: 37869901 DOI: 10.1111/dom.15331] [Citation(s) in RCA: 4] [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: 05/11/2023] [Revised: 09/12/2023] [Accepted: 09/29/2023] [Indexed: 10/24/2023]
Abstract
AIMS The objective of this umbrella review and meta-analysis was to evaluate the effect of diabetes on risk of dementia, as well as the mitigating effect of antidiabetic treatments. MATERIALS AND METHODS We conducted a systematic umbrella review on diabetes and its treatment, and a meta-analysis focusing on treatment. We searched MEDLINE/PubMed, Embase, PsycINFO, CINAHL and the Cochrane Library for systematic reviews and meta-analyses assessing the risk of cognitive decline/dementia in individuals with diabetes until 2 July 2023. We conducted random-effects meta-analyses to obtain risk ratios and 95% confidence intervals estimating the association of metformin, thiazolidinediones, pioglitazone, dipeptidyl peptidase-4 inhibitors, α-glucosidase inhibitors, meglitinides, insulin, sulphonylureas, glucagon-like peptide-1 receptor agonists (GLP1RAs) and sodium-glucose cotransporter-2 inhibitors (SGLT2is) with risk of dementia from cohort/case-control studies. The subgroups analysed included country and world region. Risk of bias was assessed with the AMSTAR tool and Newcastle-Ottawa Scale. RESULTS We included 100 reviews and 27 cohort/case-control studies (N = 3 046 661). Metformin, thiazolidinediones, pioglitazone, GLP1RAs and SGLT2is were associated with significant reduction in risk of dementia. When studies examining metformin were divided by country, the only significant effect was for the United States. Moreover, the effect of metformin was significant in Western but not Eastern populations. No significant effect was observed for dipeptidyl peptidase-4 inhibitors, α-glucosidase inhibitors, or insulin, while meglitinides and sulphonylureas were associated with increased risk. CONCLUSIONS Metformin, thiazolidinediones, pioglitazone, GLP1RAs and SGLT2is were associated with reduced risk of dementia. More longitudinal studies aimed at determining their relative benefit in different populations should be conducted.
Collapse
Affiliation(s)
- Alvin Kuate Defo
- Vascular Health Unit, Research Institute of the McGill University Health Centre, Department of Medicine, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Veselko Bakula
- Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | | | - Christopher Labos
- Vascular Health Unit, Research Institute of the McGill University Health Centre, Department of Medicine, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Simon S Wing
- Division of Endocrinology & Metabolism, Department of Medicine, McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Stella S Daskalopoulou
- Vascular Health Unit, Research Institute of the McGill University Health Centre, Department of Medicine, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- Division of Internal Medicine, Department of Medicine, McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
10
|
Sim AY, Choi DH, Kim JY, Kim ER, Goh AR, Lee YH, Lee JE. SGLT2 and DPP4 inhibitors improve Alzheimer's disease-like pathology and cognitive function through distinct mechanisms in a T2D-AD mouse model. Biomed Pharmacother 2023; 168:115755. [PMID: 37871560 DOI: 10.1016/j.biopha.2023.115755] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/17/2023] [Accepted: 10/17/2023] [Indexed: 10/25/2023] Open
Abstract
Alzheimer's disease (AD) and type 2 diabetes mellitus (T2D) share common features, including insulin resistance. Brain insulin resistance has been implicated as a key factor in the pathogenesis of AD. Recent studies have demonstrated that anti-diabetic drugs sodium-glucose cotransporter-2 inhibitor (SGLT2-i) and dipeptidyl peptidase-4 inhibitor (DPP4-i) improve insulin sensitivity and provide neuroprotection. However, the effects of these two inhibitors on the brain metabolism and insulin resistance remain uninvestigated. We developed a T2D-AD mouse model using a high-fat diet (HFD) for 19 weeks along with a single dose of streptozotocin (100 mg/kg, intraperitoneally) at the fourth week of HFD initiation. Subsequently, the animals were treated with SGLT2-i (empagliflozin, 25 mg/kg/day orally [p.o.]) and DPP4-i (sitagliptin, 100 mg/kg/day p.o.) for 7 weeks. Subsequently, behavioral tests were performed, and the expression of insulin signaling, AD-related, and other signaling pathway proteins in the brain were examined. T2D-AD mice not only showed increased blood glucose levels and body weight but also insulin resistance. SGLT2-i and DPP4-i effectively ameliorated insulin sensitivity and reduced body weight in these mice. Furthermore, SGLT2-i and DPP4-i significantly improved hippocampal-dependent learning, memory, and cognitive functions in the T2D-AD mouse model. Interestingly, SGLT2-i and DPP4-i reduced the hyperphosphorylated tau (pTau) levels and amyloid β (Aβ) accumulation and enhanced brain insulin signaling. SGLT2-i reduced pTau accumulation through the angiotensin converting enzyme-2/angiotensin (1-7)/ mitochondrial assembly receptor axis, whereas DPP4-i reduced Aβ accumulation by increasing insulin-degrading enzyme levels. These findings suggest that SGLT2-i and DPP4-i prevent AD-like pathology and cognitive dysfunction in T2D mice potentially through affecting brain insulin signaling via different mechanisms.
Collapse
Affiliation(s)
- A Young Sim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea; Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Da Hyun Choi
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Systems Biology, Glycosylation Network Research Center, Yonsei University, Seoul, Republic of Korea; Interdisciplinary Program of Integrated OMICS for Biomedical Science, Yonsei University, Seoul, Republic of Korea.
| | - Jong Youl Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Eun Ran Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - A-Ra Goh
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea; Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Yong-Ho Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Systems Biology, Glycosylation Network Research Center, Yonsei University, Seoul, Republic of Korea; Interdisciplinary Program of Integrated OMICS for Biomedical Science, Yonsei University, Seoul, Republic of Korea.
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea; Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea; Brain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
11
|
Gwizdala KL, Bazzano LA, Newton RL, Carmichael OT. Race and sex differences in the association between lifespan glycemic status and midlife cognitive function: the Bogalusa heart study. Front Public Health 2023; 11:1200415. [PMID: 38035298 PMCID: PMC10684774 DOI: 10.3389/fpubh.2023.1200415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 10/13/2023] [Indexed: 12/02/2023] Open
Abstract
Introduction Glycemic markers throughout life are associated with increased risk of midlife cognitive decline, yet it is unclear whether these associations differ by race and sex. Methods This study used cross-sectional analysis of prospectively maintained cohort. 1,295 participants in the Bogalusa Heart Study, a biracial epidemiological cohort located in a micropolitan area core setting, provided fasting plasma insulin (FPI) and glucose (FPG) biannually from 1973 to 2016. Memory, executive function (EF), attention, working memory (WM), and global cognition (GC), collected 2013-2016. Glycemic markers (i.e., FPG, FPI, and HOMA-IR) averaged within lifespan epochs (≤ 20 years, childhood/adolescence (C/A); 21-40 years, early adulthood (EA); and 40-58 years, midlife). Linear regression models were analyzed for each epoch and separate models were analyzed with sex and race, education as a covariate. Results Sample was 59% women, 34% African American (AA). Among women, higher C/A FPG was associated with poorer memory and poorer GC. Higher EA FPG was associated with poorer WM. Among men, higher EA HOMA-IR was associated with worse attention. Higher C/A HOMA-IR and FPI were associated with better memory, as was higher EA FPI. Among AA, higher C/A FPG was associated with worse attention, EF, and GC. Higher EA HOMA-IR was associated with worse attention. Higher midlife FPI and C/A HOMA-IR were associated with worse WM and EF among White Americans (WAs). Discussion Markers indicative of hyperglycemia at different epochs were associated with worse midlife cognition in women, AAs, and WAs; but not in men. Differences in the relationship between lifespan glycemic exposures and midlife cognition could reflect broader health disparities.
Collapse
Affiliation(s)
- Kathryn L. Gwizdala
- Physical Activity and Ethnic Minority Health Laboratory, Department of Population and Public Health Sciences, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
| | - Lydia A. Bazzano
- Tulane Center for Lifespan Epidemiology Research, Tulane University School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Robert L. Newton
- Physical Activity and Ethnic Minority Health Laboratory, Department of Population and Public Health Sciences, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
| | - Owen T. Carmichael
- Biomedical Imaging Center, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
| |
Collapse
|
12
|
Racsa PN, Booth TA, Chung LN, Dixon SW, Poonawalla IB. Association of medication adherence quality measures for diabetes, hypertension, and hyperlipidemia with cognitive decline. J Family Med Prim Care 2023; 12:2667-2675. [PMID: 38186814 PMCID: PMC10771214 DOI: 10.4103/jfmpc.jfmpc_935_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/19/2023] [Accepted: 08/09/2023] [Indexed: 01/09/2024] Open
Abstract
Background While diabetes, hypertension, and hyperlipidemia each are associated with increased risk of cognitive decline, little is known regarding how nonadherence to medications for these conditions is associated with cognitive decline risk. Methods We identified patients enrolled in a Medicare Advantage Prescription Drug plan who were eligible for inclusion in the CMS Star Medication Adherence quality measures for diabetes, hypertension, and hyperlipidemia in 2018, 2019, and 2020. To achieve an adherence quality measure, patients had to meet 80% of the proportion of days for the medication. We used propensity score with inverse probability of treatment weighting to balance outcomes for baseline characteristics and logistic regression models to compare odds of cognitive decline outcomes across patient groups. Results The study population of 99,774 individuals had a mean age of 71.0 years and was 49.1% female, 73.9% White, and 17.8% Black, with 62.0% living in an urban setting. Compared with patients who missed zero adherence measures, those who missed one measure had 23%-33% increased odds of cognitive decline (any decline OR = 1.23; dementia OR = 1.33; Alzheimer's disease OR = 1.27; all P values <0.01). Patients who missed 2-3 measures had 37%-96% increased odds of cognitive decline (any decline OR = 1.37; dementia OR = 1.58; Alzheimer's disease OR = 1.96; all P values <0.01). Patients who missed ≥4 adherence measures had the greatest odds of cognitive decline (any decline OR = 1.64; dementia OR = 2.05; Alzheimer's disease OR = 2.48; all P values <0.01). Conclusion Not achieving CMS Star Medication Adherence quality measures for diabetes, hypertension, and hyperlipidemia therapies was associated with increased risk of cognitive decline outcomes.
Collapse
Affiliation(s)
- Patrick N. Racsa
- Affiliated with Humana at the Time of the Work, Humana Healthcare Research, Inc., Humana Inc., 500 W Main Street Louisville, KY 40202 USA
| | - Tori A. Booth
- Humana Pharmacy Solutions, Humana Inc. 500 W Main Street Louisville, KY 40202, USA
| | - Linda N. Chung
- Humana Pharmacy Solutions, Humana Inc. 500 W Main Street Louisville, KY 40202, USA
| | - Suzanne W. Dixon
- Humana Healthcare Research, Inc., Humana Inc., 500 W Main Street Louisville, KY 40202, USA
| | - Insiya B. Poonawalla
- Humana Healthcare Research, Inc., Humana Inc., 500 W Main Street Louisville, KY 40202, USA
| |
Collapse
|
13
|
Tang H, Shao H, Shaaban CE, Yang K, Brown J, Anton S, Wu Y, Bress A, Donahoo WT, DeKosky ST, Bian J, Guo J. Newer glucose-lowering drugs and risk of dementia: A systematic review and meta-analysis of observational studies. J Am Geriatr Soc 2023; 71:2096-2106. [PMID: 36821780 PMCID: PMC10363181 DOI: 10.1111/jgs.18306] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 01/01/2023] [Accepted: 01/28/2023] [Indexed: 02/25/2023]
Abstract
BACKGROUND Preclinical studies have suggested potential beneficial effects of newer glucose-lowering drugs (GLDs) including dipeptidyl peptidase (DPP)-4 inhibitors, glucagon-like peptide-1 receptor agonists (GLP-1RAs), and sodium glucose co-transporter-2 (SGLT2) inhibitors, in protecting humans against cognitive decline and dementia. However, population studies aiming to demonstrate such cognitive benefits from newer GLDs have produced mixed findings. This meta-analysis aimed to evaluate the association between newer GLDs and risk of dementia in adults with type 2 diabetes (T2D). METHODS Electronic databases were searched up to March 11, 2022 to include observational studies that examined the association between DPP-4 inhibitors, GLP-1RAs, and SGLT2 inhibitors and risk of dementia (including all-cause dementia, Alzheimer's disease [AD], and vascular dementia [VD]) in people with T2D. We conducted a random-effects meta-analysis to calculate the relative risk (RR) with 95% confidence interval (CI) for each class of newer GLD. RESULTS Ten studies (from nine articles) involving 819,511 individuals with T2D were included. Three studies found that SGLT2 inhibitor users had a lower risk of all-cause dementia than non-SGLT2 inhibitor users (RR, 0.62; 95% CI, 0.39-0.97). Five studies found that users versus nonusers of GLP-1RAs were associated with a significant reduction in the risk of all-cause dementia (RR, 0.72; 95% CI, 0.54-0.97). However, a meta-analysis for AD and VD was unavailable for SGLT2 inhibitors and GLP-1RAs because only one study was included for each drug. In seven studies, users vs. nonusers of DPP-4 inhibitors were significantly associated with a decreased risk of all-cause dementia (RR, 0.84; 95% CI, 0.74-0.94) and VD (RR, 0.59; 95% CI, 0.47-0.75) but not AD (RR, 0.82; 95% CI, 0.63-1.08). CONCLUSION Newer GLDs were associated with a decreased risk of all-cause dementia in people with T2D. Because of the observational nature and significant heterogeneity between studies, the results should be interpreted with caution. Further research is warranted to confirm our findings.
Collapse
Affiliation(s)
- Huilin Tang
- Department of Pharmaceutical Outcomes and Policy, University of Florida College of Pharmacy, Gainesville, Florida, USA
| | - Hui Shao
- Department of Pharmaceutical Outcomes and Policy, University of Florida College of Pharmacy, Gainesville, Florida, USA
- Center for Drug Evaluation and Safety, University of Florida, Gainesville, Florida, USA
| | - C. Elizabeth Shaaban
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Keming Yang
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Joshua Brown
- Department of Pharmaceutical Outcomes and Policy, University of Florida College of Pharmacy, Gainesville, Florida, USA
- Center for Drug Evaluation and Safety, University of Florida, Gainesville, Florida, USA
| | - Stephen Anton
- Department of Aging and Geriatric Research, College of Medicine, University of Florida, Gainesville, Florida, USA
- Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, USA
| | - Yonghui Wu
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Adam Bress
- Division of Health System Innovation and Research, Department of Population Health Sciences, University of Utah, Salt Lake City, Utah, USA
| | - William T Donahoo
- Division of Endocrinology, Diabetes and Metabolism, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Steven T DeKosky
- Department of Neurology and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida USA
- 1Florida Alzheimer’s Disease Research Center (ADRC), University of Florida, Gainesville, Florida, USA
| | - Jiang Bian
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Jingchuan Guo
- Department of Pharmaceutical Outcomes and Policy, University of Florida College of Pharmacy, Gainesville, Florida, USA
- Center for Drug Evaluation and Safety, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
14
|
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: 6.0] [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
|
15
|
Sebastian MJ, Khan SKA, Pappachan JM, Jeeyavudeen MS. Diabetes and cognitive function: An evidence-based current perspective. World J Diabetes 2023; 14:92-109. [PMID: 36926658 PMCID: PMC10011899 DOI: 10.4239/wjd.v14.i2.92] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/26/2022] [Accepted: 01/16/2023] [Indexed: 02/14/2023] Open
Abstract
Several epidemiological studies have clearly identified diabetes mellitus (DM) as a major risk factor for cognitive dysfunction, and it is going to be a major public health issue in the coming years because of the alarming rise in diabetes prevalence across the world. Brain and neural tissues predominantly depend on glucose as energy substrate and hence, any alterations in carbohydrate meta-bolism can directly impact on cerebral functional output including cognition, executive capacity, and memory. DM affects neuronal function and mental capacity in several ways, some of which include hypoperfusion of the brain tissues from cerebrovascular disease, diabetes-related alterations of glucose transporters causing abnormalities in neuronal glucose uptake and metabolism, local hyper- and hypometabolism of brain areas from insulin resistance, and recurrent hypoglycemic episodes inherent to pharmacotherapy of diabetes resulting in neuronal damage. Cognitive decline can further worsen diabetes care as DM is a disease largely self-managed by patients. Therefore, it is crucial to understand the pathobiology of cognitive dysfunction in relation to DM and its management for optimal long-term care plan for patients. A thorough appraisal of normal metabolic characteristics of the brain, how alterations in neural metabolism affects cognition, the diagnostic algorithm for patients with diabetes and dementia, and the management and prognosis of patients when they have this dangerous combination of illnesses is imperative in this context. This evidence-based narrative with the back-up of latest clinical trial reviews elaborates the current understanding on diabetes and cognitive function to empower physicians to manage their patients in day-to-day clinical practice.
Collapse
Affiliation(s)
| | - Shahanas KA Khan
- Department of Endocrinology and Metabolism, Lancashire Teaching Hospitals NHS Trust, Preston PR2 9HT, United Kingdom
| | - Joseph M Pappachan
- Department of Endocrinology and Metabolism, Lancashire Teaching Hospitals NHS Trust, Preston PR2 9HT, United Kingdom
- Faculty of Science, Manchester Metropolitan University, Manchester M15 6BH, United Kingdom
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - Mohammad Sadiq Jeeyavudeen
- Department of Endocrinology and Metabolism, University Hospitals of Edinburgh, Edinburgh EH16 4SA, United Kingdom
| |
Collapse
|
16
|
Sui H, Zhu L, Zhan L, Bi T, Zhang B. ZiBuPiYin recipe ameliorates diabetes-associated cognitive decline by improving neuronal mitochondrial function in chronic psychologically stressed zucker diabetic fatty rats. JOURNAL OF ETHNOPHARMACOLOGY 2023; 302:115947. [PMID: 36403740 DOI: 10.1016/j.jep.2022.115947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 10/13/2022] [Accepted: 11/13/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zibu Piyin Recipe (ZBPYR) is a traditional Chinese medicine compound composed of 12 kinds of Chinese herbal medicines including red ginseng and yam. Long-term basic and clinical applications have proved that ZBPYR can prevent and treat cognitive dysfunction. Previous studies showed that chronic psychological stress can increase the risk of type 2 diabetes mellitus (T2DM), and lead to cognitive decline. Mitochondrial dysfunction plays a key role in chronic psychological stress-induced diabetes mellitus. While the mechanism of mitochondrial dysfunction and insulin resistance in diabetes-associated cognitive decline (DACD) is unclear. AIM OF THE STUDY Our previous research found that a ZiBuPiYin recipe (ZBPYR) has significant pharmacological effects against DACD. The present study investigated changes in mitochondrial dysfunction in the brain and the mechanism of insulin resistance and mitochondrial damage to explore the relationship between neuronal mitochondrial dysfunction and insulin resistance in chronic psychologically stressed DACD rats. MATERIALS AND METHODS Zucker diabetic fatty (ZDF) rats with spontaneous T2DM and rats with diabetic cognitive impairment that was induced by chronic psychological stress were used in in vivo experiments. PC12 cells that were damaged by rotenone were used for the in vitro experiment. RESULTS The findings indicated that the number of mitochondria decreased, morphology and membrane potential were damaged, and reactive oxygen species increased in the cortex and hippocampus in psychologically stressed DACD rats. Protein kinase Cβ2 (PKCβ2) activation and insulin resistance were markedly induced by chronic psychological stress, together with decreases in peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α) and mitochondrial fusion protein 2 (Mfn2). Furthermore, ZBPYR exerted protective effects both in in vivo and in vitro. CONCLUSION Mitochondrial damage and insulin resistance were observed in the brain in chronic psychologically stressed DACD rats. The ZBPYR significantly improved brain mitochondrial damage and insulin resistance in chronic psychologically stressed DACD rats. These results provide novel insights for the development of ZBPYR as a traditional Chinese medicine for the treatment of chronic psychological stress and DACD.
Collapse
Affiliation(s)
- Hua Sui
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 116044, Liaoning, China.
| | - Lianlian Zhu
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, China.
| | - Libin Zhan
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, China.
| | - Tingting Bi
- School of Traditional Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China.
| | - Boyu Zhang
- Institute of Integrative Medicine, Dalian Medical University, Dalian, 116044, Liaoning, China.
| |
Collapse
|
17
|
Sood A, Fernandes V, Preeti K, Khot M, Khatri DK, Singh SB. Fingolimod Alleviates Cognitive Deficit in Type 2 Diabetes by Promoting Microglial M2 Polarization via the pSTAT3-jmjd3 Axis. Mol Neurobiol 2023; 60:901-922. [PMID: 36385233 DOI: 10.1007/s12035-022-03120-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 11/03/2022] [Indexed: 11/17/2022]
Abstract
Sphingosine receptors (S1PRs) are implicated in the progression of neurodegenerative diseases and metabolic disorders like obesity and type 2 diabetes (T2D). The link between S1PRs and cognition in type 2 diabetes, as well as the mechanisms that underpin it, are yet unknown. Neuroinflammation is the common pathology shared among T2D and cognitive impairment. However, the interplay between the M1 and M2 polarization state of microglia, a primary driver of neuroinflammation, could be the driving factor for impaired learning and memory in diabetes. In the present study, we investigated the effects of fingolimod (S1PR1 modulator) on cognition in high-fat diet and streptozotocin-induced diabetic mice. We further assessed the potential pathways linking microglial polarization and cognition in T2D. Fingolimod (0.5 mg/kg and 1 mg/kg) improved M2 polarization and synaptic plasticity while ameliorating cognitive decline and neuroinflammation. Sphingolipid dysregulation was mimicked in vitro using palmitate in BV2 cells, followed by conditioned media exposure to Neuro2A cells. Mechanistically, type 2 diabetes induced microglial activation, priming microglia towards the M1 phenotype. In the hippocampus and cortex of type 2 diabetic mice, there was a substantial drop in pSTAT3, which was reversed by fingolimod. This protective effect of fingolimod on microglial M2 polarization was primarily suppressed by selective jmjd3 blockade in vitro using GSK-J4, revealing that jmjd3 was involved downstream of STAT3 in the fingolimod-enabled shift of microglia from M1 to M2 polarization state. This study suggested that fingolimod might effectively improve cognition in type 2 diabetes by promoting M2 polarization.
Collapse
Affiliation(s)
- Anika Sood
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, 500037, Hyderabad, India
| | - Valencia Fernandes
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, 500037, Hyderabad, India
| | - Kumari Preeti
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, 500037, Hyderabad, India
| | - Mayuri Khot
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, 500037, Hyderabad, India
| | - Dharmendra Kumar Khatri
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, 500037, Hyderabad, India.
| | - Shashi Bala Singh
- Molecular & Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, 500037, Hyderabad, India.
| |
Collapse
|
18
|
Dao L, Choi S, Freeby M. Type 2 diabetes mellitus and cognitive function: understanding the connections. Curr Opin Endocrinol Diabetes Obes 2023; 30:7-13. [PMID: 36385094 DOI: 10.1097/med.0000000000000783] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
PURPOSE OF REVIEW To review the connection between type 2 diabetes and cognitive dysfunction, including its epidemiology, potential mechanisms of pathophysiology, risk factors, possible prevention, and treatment considerations. RECENT FINDINGS Diabetes is a risk factor for mild cognitive decline, in addition to Alzheimer's disease and vascular dementia. Duration of diabetes, concomitant vascular or associated co-morbidities, hyper- and hypoglycemia may lead to worsening cognitive dysfunction. Unfortunately, there is a lack of evidence-based guidance on the prevention of cognitive dysfunction in the diabetes population. Studies of diabetes medications, including metformin, glucagon-like peptide-1 (GLP-1) receptor agonists, and sodium-glucose cotransporter-2 inhibitors (SGLT2) have shown some benefit with cardiovascular morbidity and may affect cognition. In the absence of clearly defined preventive tools, diabetes practice guidelines recommend annual cognitive screening as standard of care in adults with diabetes aged 65 years or older. SUMMARY People living with diabetes are at risk for significant decline in cognitive function. Epidemiology and risk factors are well defined. Prevention and treatment strategies are limited and require further study.
Collapse
Affiliation(s)
- Lisa Dao
- Division of Endocrinology, Diabetes and Metabolism, David Geffen School of Medicine UCLA
| | - Sarah Choi
- UCLA School of Nursing, Los Angeles, California, USA
| | - Matthew Freeby
- Division of Endocrinology, Diabetes and Metabolism, David Geffen School of Medicine UCLA
| |
Collapse
|
19
|
Lin Y, Gong Z, Ma C, Wang Z, Wang K. Relationship between glycemic control and cognitive impairment: A systematic review and meta-analysis. Front Aging Neurosci 2023; 15:1126183. [PMID: 36776436 PMCID: PMC9909073 DOI: 10.3389/fnagi.2023.1126183] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 01/11/2023] [Indexed: 01/27/2023] Open
Abstract
Background Diabetes mellitus, or hyperglycemia, is an independent risk factor for cognitive impairment. Here we systematically analyzed whether glycemic control could improve cognitive impairment in patients with diabetes mellitus (DM), hyperglycemia, or insulin resistance. Methods Three databases (PubMed, EMBASE, and Cochrane Library) and ClinicalTrials.gov were searched for randomized controlled trials analyzing the relationship between glycemic control and cognitive function assessments, published from database inception to June 2022. Patients in experimental groups were treated with antidiabetic drugs, while control groups were treated with a placebo or alternative antidiabetic drugs. Data analysis was conducted using RevMan 5.3 and StataSE-64, and standardized mean difference (SMD) and 95% confidence intervals (CIs) were calculated. Results Thirteen studies comprising 19,314 participants were included. Analysis revealed that glycemic control significantly attenuated the degree of decline in cognitive function assessment scores (SMD = 0.15; 95% CI 0.05, 0.26; p < 0.00001), and funnel plots confirmed no publication bias. Seven studies used Mini-Mental State Examination as the primary cognitive function assessment, showing that glycemic control significantly delayed the degree of decline in cognitive function assessment scores (SMD = 0.18; 95% CI 0.03, 0.34; p = 0.02). Similar results were seen in two studies using the Montreal Cognitive Assessment scale, but without significant difference (SMD = 0.05; 95% CI-0.10, 0.21; p = 0.51). One study using Auditory Word Learning Test (AVLT) showed that glycemic control significantly delayed the decline in cognitive function assessment scores (SMD = 0.52; 95% CI 0.11,0.93; p = 0.01), and another used Wechsler Memory Scale Revised, showing similar results (SMD = 1.45; 95% CI 0.86, 2.04; p < 0.00001). Likewise, a study that used Modified Mini-Mental State scale showed that glycemic control significantly delayed the decline in cognitive function assessment scores (SMD = -0.10; 95% CI-0.16, -0.03; p = 0.005). Lastly, one study used AVLT subtests to show that glycemic control delayed the decline in cognitive function assessment scores, although not statistically significant (SMD = 0.09; 95% CI-0.53, 0.71; p = 0.78). Conclusion Glycemic control through antidiabetic treatment correlates with the improvement of cognitive impairment in patients with DM, hyperglycemia or insulin resistance. However, further studies are needed to validate the results of this study. Systematic Review Registration PROSPERO, identifier CRD42022342260.
Collapse
Affiliation(s)
- Yufeng Lin
- Department of Neurology, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Zhongying Gong
- Department of Neurology, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Chunchao Ma
- Department of Neurology, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Zhiyun Wang
- Department of Neurology, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China,*Correspondence: Kaiyuan Wang, ; Zhiyun Wang,
| | - Kaiyuan Wang
- Department of Anesthesiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China,*Correspondence: Kaiyuan Wang, ; Zhiyun Wang,
| |
Collapse
|
20
|
Ji X, Gao H, Sun D, Zhuang J, Fang Y, Wang K, Ahmadizar F. Trajectories of Cognition and Daily Functioning Before and After Incident Diabetes. Diabetes Care 2023; 46:75-82. [PMID: 36378879 DOI: 10.2337/dc22-1190] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 10/09/2022] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The temporal pattern of cognitive and functional change before and after incident diabetes remains unknown. RESEARCH DESIGN AND METHODS Data from wave 2 to wave 9 (2004-2018) of the English Longitudinal Study of Ageing were used. Global cognition (assessed by orientation, memory, and executive function) and daily functioning (calculated as the sum of impaired basic and instrumental activities of daily living) were measured in each wave. Incident diabetes was defined as glycated hemoglobin A1c ≥6.5% (47.5 mmol/mol), self-reported doctor diagnosis of diabetes, or glucose-lowering medication use during follow-up. RESULTS Among the 6,342 participants (mean age 65.0 years, 57.8% women) included, 576 participants (9.1%) with incident diabetes were identified during a median follow-up of 13.3 years. The annual rates of change in global cognition (β = -0.035 SD/year; 95% CI -0.054 to -0.015), orientation (-0.031 SD/year; -0.060 to -0.002), memory (-0.016 SD/year; -0.029 to -0.003), and executive function (-0.027 SD/year; -0.042 to -0.013) were accelerated after diabetes diagnosis compared with before the event. The postdiabetes annual changes in daily functioning (0.093 points/year; 95% CI 0.056-0.131) were also accelerated compared with the prediabetes diagnosis. However, the rate of cognitive and functional decline before the diabetes diagnosis in participants with future incident diabetes was similar to the rate in participants without diabetes. Also, no significant acute change was observed during its onset. CONCLUSIONS Incident diabetes is associated with accelerated cognitive and functional decline after, but not before, the event. We suggest careful monitoring for cognitive and physical dysfunction after a diabetes diagnosis.
Collapse
Affiliation(s)
- Xiaoli Ji
- Department of Occupational Disease, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hui Gao
- Changning Center for Disease Control and Prevention, Shanghai, China
| | - Daoyuan Sun
- Department of Occupational Disease, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianlin Zhuang
- Changning Center for Disease Control and Prevention, Shanghai, China
| | - Yuan Fang
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Kan Wang
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Fariba Ahmadizar
- Department of Data Science and Biostatistics, Julius Global Health, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
21
|
Nagar SD, Pemu P, Qian J, Boerwinkle E, Cicek M, Clark CR, Cohn E, Gebo K, Loperena R, Mayo K, Mockrin S, Ohno-Machado L, Ramirez AH, Schully S, Able A, Green A, Zuchner S, Jordan IK, Meller R. Investigation of hypertension and type 2 diabetes as risk factors for dementia in the All of Us cohort. Sci Rep 2022; 12:19797. [PMID: 36396674 PMCID: PMC9672061 DOI: 10.1038/s41598-022-23353-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 10/31/2022] [Indexed: 11/18/2022] Open
Abstract
The World Health Organization recently defined hypertension and type 2 diabetes (T2D) as modifiable comorbidities leading to dementia and Alzheimer's disease. In the United States (US), hypertension and T2D are health disparities, with higher prevalence seen for Black and Hispanic minority groups compared to the majority White population. We hypothesized that elevated prevalence of hypertension and T2D risk factors in Black and Hispanic groups may be associated with dementia disparities. We interrogated this hypothesis using a cross-sectional analysis of participant data from the All of Us (AoU) Research Program, a large observational cohort study of US residents. The specific objectives of our study were: (1) to compare the prevalence of dementia, hypertension, and T2D in the AoU cohort to previously reported prevalence values for the US population, (2) to investigate the association of hypertension, T2D, and race/ethnicity with dementia, and (3) to investigate whether race/ethnicity modify the association of hypertension and T2D with dementia. AoU participants were recruited from 2018 to 2019 as part of the initial project cohort (R2019Q4R3). Participants aged 40-80 with electronic health records and demographic data (age, sex, race, and ethnicity) were included for analysis, yielding a final cohort of 125,637 individuals. AoU participants show similar prevalence of hypertension (32.1%) and T2D (13.9%) compared to the US population (32.0% and 10.5%, respectively); however, the prevalence of dementia for AoU participants (0.44%) is an order of magnitude lower than seen for the US population (5%). AoU participants with dementia show a higher prevalence of hypertension (81.6% vs. 31.9%) and T2D (45.9% vs. 11.4%) compared to non-dementia participants. Dominance analysis of a multivariable logistic regression model with dementia as the outcome shows that hypertension, age, and T2D have the strongest associations with dementia. Hispanic was the only race/ethnicity group that showed a significant association with dementia, and the association of sex with dementia was non-significant. The association of T2D with dementia is likely explained by concurrent hypertension, since > 90% of participants with T2D also had hypertension. Black race and Hispanic ethnicity interact with hypertension, but not T2D, to increase the odds of dementia. This study underscores the utility of the AoU participant cohort to study disease prevalence and risk factors. We do notice a lower participation of aged minorities and participants with dementia, revealing an opportunity for targeted engagement. Our results indicate that targeting hypertension should be a priority for risk factor modifications to reduce dementia incidence.
Collapse
Affiliation(s)
| | - Priscilla Pemu
- Morehouse School of Medicine, Atlanta, USA
- University of Miami, Coral Gables, USA
| | - Jun Qian
- All of Us Demonstration Projects Subcommittee, National Institutes of Health, Bethesda, USA
- Vanderbilt University Medical Center, Nashville, USA
| | - Eric Boerwinkle
- All of Us Demonstration Projects Subcommittee, National Institutes of Health, Bethesda, USA
- The University of Texas Health Science Center at Houston, Houston, USA
| | - Mine Cicek
- All of Us Demonstration Projects Subcommittee, National Institutes of Health, Bethesda, USA
- Mayo Clinic, Rochester, USA
| | - Cheryl R Clark
- All of Us Demonstration Projects Subcommittee, National Institutes of Health, Bethesda, USA
- Brigham and Women's Hospital, Boston, USA
| | - Elizabeth Cohn
- All of Us Demonstration Projects Subcommittee, National Institutes of Health, Bethesda, USA
- Hunter College, New York, USA
| | - Kelly Gebo
- All of Us Demonstration Projects Subcommittee, National Institutes of Health, Bethesda, USA
- Johns Hopkins University, Baltimore, USA
| | - Roxana Loperena
- All of Us Demonstration Projects Subcommittee, National Institutes of Health, Bethesda, USA
- Vanderbilt University Medical Center, Nashville, USA
| | - Kelsey Mayo
- All of Us Demonstration Projects Subcommittee, National Institutes of Health, Bethesda, USA
- Vanderbilt University Medical Center, Nashville, USA
| | - Stephen Mockrin
- All of Us Demonstration Projects Subcommittee, National Institutes of Health, Bethesda, USA
- National Institutes of Health, Bethesda, USA
| | - Lucila Ohno-Machado
- All of Us Demonstration Projects Subcommittee, National Institutes of Health, Bethesda, USA
- University of California, San Diego, USA
| | - Andrea H Ramirez
- All of Us Demonstration Projects Subcommittee, National Institutes of Health, Bethesda, USA
- Vanderbilt University Medical Center, Nashville, USA
| | - Sheri Schully
- All of Us Demonstration Projects Subcommittee, National Institutes of Health, Bethesda, USA
- Vanderbilt University Medical Center, Nashville, USA
| | - Ashley Able
- All of Us Demonstration Projects Subcommittee, National Institutes of Health, Bethesda, USA
- Vanderbilt University Medical Center, Nashville, USA
| | - Ashley Green
- All of Us Demonstration Projects Subcommittee, National Institutes of Health, Bethesda, USA
| | - Stephan Zuchner
- Vanderbilt University Medical Center, Nashville, USA
- University of Miami, Coral Gables, USA
| | | | - Robert Meller
- Morehouse School of Medicine, Atlanta, USA.
- University of Miami, Coral Gables, USA.
| |
Collapse
|
22
|
Wang Q, Li X, Teng N, Li X. Prediabetes and the incidence of dementia in general population: a systematic review and meta-analysis of prospective studies. Psychogeriatrics 2022; 22:666-678. [PMID: 35777977 DOI: 10.1111/psyg.12869] [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: 04/13/2022] [Revised: 05/23/2022] [Accepted: 06/01/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Continuous hyperglycaemia has been related with dementia. However, it remains unclear whether prediabetes poses a higher risk of dementia. A meta-analysis was therefore conducted to comprehensively investigate the possible role of prediabetes as a risk factor of dementia. METHODS Prospective cohort studies reporting the association of prediabetes and dementia were identified from PubMed, Web of Science, and Embase databases. A random-effects model was applied to combine the results by incorporating the influence of heterogeneity. Subgroup analyses were also conducted to explore the influences of study features on the relationship. Sensitivity analysis re-estimated the combined effect size after excluding single studies separately to explore the robustness of the results. RESULTS Nine studies involving 29 986 adults from the general population, 6265 (20.9%) of whom had prediabetes, were included. It was shown that prediabetes was not independently associated with a higher incidence of dementia compared with normoglycaemia (adjusted risk ratio (RR): 1.01, 95% confidence interval (CI): 0.85-1.21, P = 0.89, I2 = 39%). Subgroup analyses according to the definitions of prediabetes, follow-up duration, method for diagnosis of dementia, and quality score produced similar findings (P for all subgroup differences >0.05). In addition, prediabetes was not independently associated with the incidence of Alzheimer's disease (RR: 1.24, 95% CI: 0.98-1.56, P = 0.07, I2 = 0%) or vascular dementia (RR: 1.16, 95% CI: 0.70-1.92, P = 0.56, I2 = 0%). Different definitions of prediabetes have the potential to influence the results, as reflected in the subgroup analysis for Alzheimer's disease (RR: 1.30, 95% CI: 1.06-1.60, P = 0.01, I2 = 0%). CONCLUSIONS Prediabetes may not be an independent risk factor of all-cause dementia or vascular dementia in the general adult population. However, changing the definition of prediabetes may have an impact on the outcome for Alzheimer's disease.
Collapse
Affiliation(s)
- Qihao Wang
- The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xinying Li
- The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Nan Teng
- School of Public Health, Dalian Medical University, Dalian, China
| | - Xiaofeng Li
- School of Public Health, Dalian Medical University, Dalian, China
| |
Collapse
|
23
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
|
24
|
Singh A, Bodakhe SH. Resveratrol attenuates behavioural impairment associated with learning and memory in HFD-STZ induced diabetic rats. Br J Pharmacol 2022; 179:4673-4691. [PMID: 35710260 DOI: 10.1111/bph.15895] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/10/2022] [Accepted: 04/22/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Literature have indicated that a high-fat diet (HFD) is a common risk factor for type 2 diabetes mellitus (T2DM) and its associated cognitive-impairments. Mounting evidence supports that, in the diabetic animal model, resveratrol (RSV, SIRT1-modulator) can regulate the fasting glucose and antioxidant levels, as well as the lipid profile, and may alleviate the cognitive-dysfunction associated with diabetes. EXPERIMENTAL APPROACH Albino rats were fed 60% HFD-STZ (45mg/kg,i.p, single dose) to induce T2DM so that the experimental T2DM animal model could be used. After 14 weeks of the animals being in a confirmed diabetic condition, they were divided into various groups and treated with metformin(200mg/kg,i.p.) and RSV(50 and 100 mg/kg,i.p.) for four weeks. A multimodal approach involving oxidative-nitroso-stress, SIRT1, TGF-β1 levels, inflammation, cholinergic activity (serum, hippocampus, cerebral cortex), and a battery of behavioural studies associated with learning-memory were performed during and after the experimental-protocol. KEY RESULTS The administration of RSV significantly attenuated the increased glucose levels (pre, and post-prandial), impaired glucose tolerance, HbA1c, and decreased the body weights of the T2DM rats. Moreover, RSV ameliorated the impaired learning and memory-associated with increased SIRT1 and the decreased TGF-β1, TNF-α, oxidative-nitroso-stress and cholinergic activities in the serum and the brains of the T2DM-animals. CONCLUSION AND IMPLICATION Our investigations demonstrate that SIRT1-modulation can inter-play with TGF-β1 signalling, as well as mitigate hyperglycaemia and subsequent learning-memory impairments, in the T2DM-animals. Moreover, our study showed that novel therapeutic-targets, including TGF-β1, may add to our knowledge of RSV when used in the treatment of impaired memory-associated with diabetes.
Collapse
Affiliation(s)
- Amrita Singh
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India.,Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Surendra H Bodakhe
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| |
Collapse
|
25
|
Colca JR, Finck BN. Metabolic Mechanisms Connecting Alzheimer's and Parkinson's Diseases: Potential Avenues for Novel Therapeutic Approaches. Front Mol Biosci 2022; 9:929328. [PMID: 35782864 PMCID: PMC9243557 DOI: 10.3389/fmolb.2022.929328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's (AD) and Parkinson's Diseases (PD) are common neurodegenerative disorders growing in incidence and prevalence and for which there are no disease-modifying treatments. While there are considerable complexities in the presentations of these diseases, the histological pictures of these pathologies, as well as several rare genetic predispositions for each, point to the involvement of maladaptive protein processing and inflammation. Importantly, the common presentations of AD and PD are connected to aging and to dysmetabolism, including common co-diagnosis of metabolic syndrome or diabetes. Examination of anti-diabetic therapies in preclinical models and in some observational clinical studies have suggested effectiveness of the first generation insulin sensitizer pioglitazone in both AD and PD. Recently, the mitochondrial pyruvate carrier (MPC) was shown to be a previously unrecognized target of pioglitazone. New insulin sensitizers are in development that can be dosed to full engagement of this previously unappreciated mitochondrial target. Here we review molecular mechanisms that connect modification of pyruvate metabolism with known liabilities of AD and PD. The mechanisms involve modification of autophagy, inflammation, and cell differentiation in various cell types including neurons, glia, macrophages, and endothelium. These observations have implications for the understanding of the general pathology of neurodegeneration and suggest general therapeutic approaches to disease modification.
Collapse
Affiliation(s)
- Jerry R. Colca
- Metabolic Solutions Development Company, Western Michigan University, Kalamazoo, MI, United States
| | - Brian N. Finck
- Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
26
|
Rhee EJ. Extra-Glycemic Effects of Anti-Diabetic Medications: Two Birds with One Stone? Endocrinol Metab (Seoul) 2022; 37:415-429. [PMID: 35798548 PMCID: PMC9262696 DOI: 10.3803/enm.2022.304] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/07/2022] [Accepted: 06/12/2022] [Indexed: 11/16/2022] Open
Abstract
The world is suffering from a rapid increase in the number of people with diabetes due to the increased prevalence of obesity and lengthened life span. Since the development of insulin thanks to the efforts of Prof. Banting and Dr. Best in 1922, for which they won the Nobel Prize, remarkable developments in anti-diabetic medications have dramatically lengthened the lifespan of patients with diabetes. However, the control rate of hyperglycemia in patients with diabetes remains unsatisfactory, since glycemic control requires both medication and lifestyle modifications to slow the deterioration of pancreatic beta-cell function and prevent diabetic complications. From the initial "triumvirate" to the "ominous octet," and now the "egregious eleven," the number of organs recognized as being involved in hyperglycemia and diabetes has increased with the development of anti-diabetic medications. Recent unexpected results from outcome trials of anti-diabetic medications have enabled anti-diabetic medications to be indicated for the prevention of chronic kidney disease and heart failure, even in patients without diabetes. In this review, I would like to summarize the extra-glycemic effects of anti-diabetic medications.
Collapse
Affiliation(s)
- Eun-Jung Rhee
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
27
|
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: 0] [Impact Index Per Article: 0] [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
|
28
|
Reinke C, Buchmann N, Fink A, Tegeler C, Demuth I, Doblhammer G. Diabetes duration and the risk of dementia: a cohort study based on German health claims data. Age Ageing 2022; 51:6454655. [PMID: 34923587 PMCID: PMC8753043 DOI: 10.1093/ageing/afab231] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 09/14/2021] [Indexed: 01/21/2023] Open
Abstract
Objective Diabetes is a risk factor for dementia but little is known about the impact of diabetes duration on the risk of dementia. We investigated the effect of type 2 diabetes duration on the risk of dementia. Design Prospective cohort study using health claims data representative for the older German population. The data contain information about diagnoses and medical prescriptions from the in- and outpatient sector. Methods We performed piecewise exponential models with a linear and a quadratic term for time since first type 2 diabetes diagnosis to predict the dementia risk in a sample of 13,761 subjects (2,558 dementia cases) older than 65 years. We controlled for severity of diabetes using the Adopted Diabetes Complications Severity Index. Results We found a U-shaped dementia risk over time. After type 2 diabetes diagnosis the dementia risk decreased (26% after 1 year) and reached a minimum at 4.75 years, followed by an increase through the end of follow-up. The pattern was consistent over different treatment groups, with the strongest U-shape for insulin treatment and for those with diabetes complications at the time of diabetes diagnosis. Conclusions We identified a non-linear association of type 2 diabetes duration and the risk of dementia. Physicians should closely monitor cognitive function in diabetic patients beyond the first few years after diagnosis, because the later increase in dementia occurred in all treatment groups.
Collapse
Affiliation(s)
- Constantin Reinke
- Institute for Sociology and Demography, University of Rostock, 18057 Rostock, Germany
| | - Nikolaus Buchmann
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolic Diseases (including Division of Lipid Metabolism), Biology of Aging working group, 13353 Berlin, Germany
| | - Anne Fink
- German Center for Neurodegenerative Diseases, 53127 Bonn, Germany
| | - Christina Tegeler
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolic Diseases (including Division of Lipid Metabolism), Biology of Aging working group, 13353 Berlin, Germany
- MSB Medical School Berlin, Department of Psychology, 14197 Berlin, Germany
| | - Ilja Demuth
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolic Diseases (including Division of Lipid Metabolism), Biology of Aging working group, 13353 Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BCRT - Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany
| | - Gabriele Doblhammer
- Institute for Sociology and Demography, University of Rostock, 18057 Rostock, Germany
- German Center for Neurodegenerative Diseases, 53127 Bonn, Germany
| |
Collapse
|
29
|
Arshad F, MM S, Paplikar A, Rajendran S, Kalkonde Y, Alladi S. Vascular cognitive impairment in India: Challenges and opportunities for prevention and treatment. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2021; 3:100034. [PMID: 36324418 PMCID: PMC9616277 DOI: 10.1016/j.cccb.2021.100034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/22/2021] [Accepted: 11/23/2021] [Indexed: 06/16/2023]
Abstract
The burden of vascular contribution to cognitive impairment and dementia is substantially high in India. There are approximately 5.3 million dementia patients in India and nearly 40% are estimated to be due to vascular dementia. Several factors pose unique challenges to reducing the burden of vascular dementia and vascular cognitive impairment (VCI) in India. Wide heterogeneity in vascular risk factor profile, diversity in socioeconomic, ethnic and dietary factors, as well as regional and rural-urban differences impact uniform implementation of preventive and therapeutic strategies. There is limited evidence on the natural history of vascular disease from longitudinal cohorts in India. Additionally, the lack of advanced brain imaging and genetic information pose challenges to understanding pathophysiology and treatment response to VCI in India. Efforts are now being made to implement programmes to reduce cardiovascular risk and VCI at the population level. Cognitive and functional measures appropriate to the diverse linguistic and educational context have been developed to diagnose VCI across India. Multicentric clinical and research cohorts of stroke are also being established. Filling research gaps and developing intervention strategies for the Indian context are crucial to address the growing burden of VCI.
Collapse
Affiliation(s)
- Faheem Arshad
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Samim MM
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Avanthi Paplikar
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Srijithesh Rajendran
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | | | - Suvarna Alladi
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| |
Collapse
|
30
|
Hallam B, Rees J, Petersen I, Cooper C, Avgerinou C, Walters K. How are people with mild cognitive impairment or subjective memory complaints managed in primary care? A systematic review. Fam Pract 2021; 38:669-683. [PMID: 33907811 PMCID: PMC8604277 DOI: 10.1093/fampra/cmab014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Primary care is typically the first point of contact in the health care system for people raising concerns about their memory. However, there is still a lack of high-quality evidence and understanding about how primary care professionals (PCPs) currently manage people at higher risk of developing dementia. OBJECTIVES To systematically review management strategies provided by PCPs to reduce cognitive decline in people with mild cognitive impairment and subjective memory complaints. METHOD A systematic search for studies was conducted in December 2019 across five databases (EMBASE, Medline, PsycInfo, CINAHL and Web of Science). Methodological quality of included studies was independently assessed by two authors using the Mixed Methods Appraisal Tool. RESULTS An initial 11 719 were found, 7250 were screened and 9 studies were included in the review. Most studies were self-reported behaviour surveys. For non-pharmacological strategies, the most frequent advice PCPs provided was to increase physical activity, cognitive stimulation, diet and social stimulation. For pharmacological strategies, PCPs would most frequently not prescribe any treatment. If PCPs did prescribe, the most frequent prescriptions targeted vascular risk factors to reduce the risk of further cognitive decline. CONCLUSION PCPs reported that they are much more likely to provide non-pharmacological strategies than pharmacological strategies in line with guidelines on preventing the onset of dementia. However, the quality of evidence within the included studies is low and relies on subjective self-reported behaviours. Observational research is needed to provide an accurate reflection of how people with memory problems are managed in primary care.
Collapse
Affiliation(s)
- Brendan Hallam
- UCL Research Department of Primary Care and Population Health, London, UK
| | - Jessica Rees
- Division of Psychiatry, University College London, London, UK
| | - Irene Petersen
- UCL Research Department of Primary Care and Population Health, London, UK
| | - Claudia Cooper
- Division of Psychiatry, University College London, London, UK
| | | | - Kate Walters
- UCL Research Department of Primary Care and Population Health, London, UK
| |
Collapse
|
31
|
Zhang H, Zhu W, Niu T, Wang Z, An K, Cao W, Shi J, Wang S. Inverted U-shaped correlation between serum low-density lipoprotein cholesterol levels and cognitive functions of patients with type 2 diabetes mellitus. Lipids Health Dis 2021; 20:103. [PMID: 34511118 PMCID: PMC8436464 DOI: 10.1186/s12944-021-01534-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/30/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Low-density lipoprotein cholesterol (LDL-C) metabolic disorder is common in individuals with diabetes. The role of LDL-C in mild cognitive impairment (MCI) remains to be explored. We aim to investigate the associations between LDL-C at different levels and details of cognition decline in patients with type 2 diabetes mellitus (T2DM). METHODS Patients with T2DM (n = 497) were recruited. Clinical parameters and neuropsychological tests were compared between patients with MCI and controls. Goodness of fit was assessed to determine the linear or U-shaped relationship between LDL-C and cognitive function. The cut-off point of LDL-C was calculated. Correlation and regression were carried out to explore the relationship between cognitive dysfunction and LDL-C levels above and below the cut-off point. RESULTS Although no significant difference in LDL-C levels was detected in 235 patients with MCI, compared with 262 patients without MCI, inverted-U-shaped association was determined between LDL-C and Montreal Cognitive Assessment (MoCA). The cut-off point of LDL-C is 2.686 mmol/l. LDL-C (>2.686 mmol/l) is positively related to Trail Making Test B (TMTB) indicating executive function. LDL-C (<2.686 mmol/l) is positively associated with Clock Drawing Test (CDT) reflecting visual space function in patients with T2DM. CONCLUSION Inverted U-shaped correlation was found between serum LDL-C and cognitive function in patients with T2DM. Despite that the mechanisms of different LDL-C levels involved in special cognitive dysfunctions remain incompletely clarified, excessive LDL-C damages executive function, while the deficient LDL-C impairs visual space function. TRIAL REGISTRATION ChiCTR-OCC-15006060 .
Collapse
Affiliation(s)
- Haoqiang Zhang
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China.,School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Wenwen Zhu
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China.,School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Tong Niu
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China.,School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Zheng Wang
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China.,School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Ke An
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China.,School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Wuyou Cao
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China.,School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Jijing Shi
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China.,School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Shaohua Wang
- Department of Endocrinology, Affiliated Zhongda Hospital of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China. .,School of Medicine, Southeast University, Nanjing, 210009, PR China.
| |
Collapse
|
32
|
Soldevila-Domenech N, Forcano L, Vintró-Alcaraz C, Cuenca-Royo A, Pintó X, Jiménez-Murcia S, García-Gavilán JF, Nishi SK, Babio N, Gomis-González M, Corella D, Sorlí JV, Fernandez-Carrión R, Martínez-González MÁ, Marti A, Salas-Salvadó J, Castañer O, Fernández-Aranda F, Torre RDL. Interplay between cognition and weight reduction in individuals following a Mediterranean Diet: Three-year follow-up of the PREDIMED-Plus trial. Clin Nutr 2021; 40:5221-5237. [PMID: 34474192 DOI: 10.1016/j.clnu.2021.07.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Some cognitive profiles might facilitate successful weight loss and its maintenance. Also, weight reductions may result in cognitive benefits. However, little work to date has examined the interactions between cognition and weight changes in the context of interventions with the Mediterranean diet (MedDiet). We studied the within-subject longitudinal relationships between cognition, body mass index (BMI), physical activity (PA), and quality of life (QoL), in older adults following a MedDiet. METHODS The PREDIMED-Plus is a primary prevention trial testing the effect of a lifestyle intervention program with an energy-restricted MedDiet (er-MedDiet), weight-loss goals and PA promotion on cardiovascular disease. The PREDIMED-Plus-Cognition sub-study included 487 participants (50% women, mean age 65.2 ± 4.7 years), with overweight/obesity, metabolic syndrome and normal cognitive performance at baseline. A comprehensive neurocognitive test battery was administered at baseline and after 1 and 3 years. RESULTS Baseline higher performance in verbal memory (OR = 1.5; 95%CI 1.0, 2.1), visuoconstructive praxis and attention (OR = 1.5; 95%CI 0.9, 2.3), and inhibition (OR = 1.3; 95%CI 0.9, 1.9) were associated with a higher odd of achieving at least 8% weight loss after 3 years follow-up in participants randomized to the intervention group. There were moderate improvements in specific tests of memory and executive functions during follow-up. Higher adherence to the er-MedDiet was associated with greater improvements in memory. Women exhibited lower rates of change in global cognition, PA and QoL. Moreover, improvements in memory correlated with reductions in BMI after 1 year (βSTD = -0.14) and with improvements in PA after 3 years (βSTD = 0.13). Finally, participants who experienced greater improvements in executive functions and global cognition also experienced greater improvements in their QoL. CONCLUSIONS This study refines the understanding of the determinants and mutual interrelationships between longitudinally-assessed cognitive performance and weight loss, adding further evidence to the cognitive benefits associated with better adherence to a MedDiet. Our results also suggest that weight loss interventions tailored to the cognitive profile and gender of participants are promising avenues for future studies.
Collapse
Affiliation(s)
- Natalia Soldevila-Domenech
- Integrative Pharmacology and Systems Neurosciences Research Group, Neurosciences Research Program, Hospital Del Mar Medical Research Institute (IMIM), 08003, Barcelona, Spain; Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003, Barcelona, Spain.
| | - Laura Forcano
- Integrative Pharmacology and Systems Neurosciences Research Group, Neurosciences Research Program, Hospital Del Mar Medical Research Institute (IMIM), 08003, Barcelona, Spain; CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Cristina Vintró-Alcaraz
- CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029, Madrid, Spain; Department of Psychiatry, University Hospital of Bellvitge-IDIBELL, Barcelona, Spain; Psychiatry and Mental Health Group, Neuroscience Program, Institut D'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain.
| | - Aida Cuenca-Royo
- Integrative Pharmacology and Systems Neurosciences Research Group, Neurosciences Research Program, Hospital Del Mar Medical Research Institute (IMIM), 08003, Barcelona, Spain.
| | - Xavier Pintó
- CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029, Madrid, Spain; Lipid Unit, Department of Internal Medicine, Bellvitge Biomedical Research Institute (IDIBELL)-Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, 08908, Barcelona, Spain.
| | - Susana Jiménez-Murcia
- CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029, Madrid, Spain; Department of Psychiatry, University Hospital of Bellvitge-IDIBELL, Barcelona, Spain; Psychiatry and Mental Health Group, Neuroscience Program, Institut D'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain; Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.
| | - Jesús F García-Gavilán
- CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029, Madrid, Spain; Universitat Rovira I Virgili, Departament de Bioquímica I Biotecnologia, Unitat de Nutrició Humana, Reus, Spain; Institut D'Investigació Sanitària Pere Virgili (IISPV). Hospital Universitari San Joan de Reus, Reus, Spain.
| | - Stephanie K Nishi
- CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029, Madrid, Spain; Universitat Rovira I Virgili, Departament de Bioquímica I Biotecnologia, Unitat de Nutrició Humana, Reus, Spain; Institut D'Investigació Sanitària Pere Virgili (IISPV). Hospital Universitari San Joan de Reus, Reus, Spain.
| | - Nancy Babio
- CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029, Madrid, Spain; Universitat Rovira I Virgili, Departament de Bioquímica I Biotecnologia, Unitat de Nutrició Humana, Reus, Spain; Institut D'Investigació Sanitària Pere Virgili (IISPV). Hospital Universitari San Joan de Reus, Reus, Spain.
| | - Maria Gomis-González
- Integrative Pharmacology and Systems Neurosciences Research Group, Neurosciences Research Program, Hospital Del Mar Medical Research Institute (IMIM), 08003, Barcelona, Spain.
| | - Dolores Corella
- CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029, Madrid, Spain; Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010, Valencia, Spain.
| | - Jose V Sorlí
- CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029, Madrid, Spain; Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010, Valencia, Spain.
| | - Rebeca Fernandez-Carrión
- CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029, Madrid, Spain; Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010, Valencia, Spain.
| | - Miguel Ángel Martínez-González
- CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029, Madrid, Spain; University of Navarra, Department of Preventive Medicine and Public Health, Pamplona, Spain; Navarra's Health Research Institute (IdiSNA), Pamplona, Spain; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Amelia Marti
- CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029, Madrid, Spain; Navarra's Health Research Institute (IdiSNA), Pamplona, Spain; University of Navarra, Department of Nutrition, Food Sciences and Physiology, Pamplona, Spain.
| | - Jordi Salas-Salvadó
- CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029, Madrid, Spain; Universitat Rovira I Virgili, Departament de Bioquímica I Biotecnologia, Unitat de Nutrició Humana, Reus, Spain; Institut D'Investigació Sanitària Pere Virgili (IISPV). Hospital Universitari San Joan de Reus, Reus, Spain.
| | - Olga Castañer
- CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029, Madrid, Spain; Cardiovascular Risk and Nutrition Research Group, Hospital Del Mar Medical Research Institute (IMIM), 08003, Barcelona, Spain; Endocrinology Service. Institut Hospital Del Mar D'Investigacions Mèdiques (IMIM), 08003, Barcelona, Spain.
| | - Fernando Fernández-Aranda
- CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029, Madrid, Spain; Department of Psychiatry, University Hospital of Bellvitge-IDIBELL, Barcelona, Spain; Psychiatry and Mental Health Group, Neuroscience Program, Institut D'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain; Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.
| | - Rafael de la Torre
- Integrative Pharmacology and Systems Neurosciences Research Group, Neurosciences Research Program, Hospital Del Mar Medical Research Institute (IMIM), 08003, Barcelona, Spain; Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003, Barcelona, Spain; CIBER Fisiopatología Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| |
Collapse
|
33
|
Gong J, Harris K, Hackett M, Peters SAE, Brodaty H, Cooper M, Hamet P, Harrap S, Mancia G, MacMahon S, Chalmers J, Woodward M. Sex differences in risk factors for cognitive decline and dementia, including death as a competing risk, in individuals with diabetes: Results from the ADVANCE trial. Diabetes Obes Metab 2021; 23:1775-1785. [PMID: 33783955 DOI: 10.1111/dom.14391] [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: 01/24/2021] [Revised: 03/16/2021] [Accepted: 03/25/2021] [Indexed: 12/18/2022]
Abstract
AIM To estimate the associations between risk factors and cognitive decline (CD)/dementia, and the sex differences in these risk factors in individuals with type 2 diabetes, while accounting for the competing risk of death. MATERIALS AND METHODS The Action in Diabetes and Vascular Disease: Preterax and Diamicron Modified Release Controlled Evaluation (ADVANCE) trial of 11,140 individuals with type 2 diabetes was used to estimate the odds of CD/dementia using multinomial logistic regression. RESULTS During a median 5-year follow-up, 1827 participants (43.2% women) had CD/dementia (1718 with CD only; 21 with dementia only; 88 with CD and dementia), and 929 (31.0% women) died without CD/dementia. Women had lower odds of CD/dementia than men (odds ratio [OR] [95% confidence interval], 0.88 [0.77, 1.00]); older age, higher total cholesterol, HbA1c, waist circumference, waist-to-height ratio, moderately increased albumin-creatinine ratio, stroke/transient ischaemic attack and retinal disease were each associated with greater odds of CD/dementia; higher years at education completion, baseline cognitive function, taller stature and current alcohol use were inversely associated. Higher waist circumference (women-to-men ratio of ORs [ROR], 1.05 [1.00, 1.10] per 5 cm) and presence of anxiety/depression (ROR, 1.28 [1.01, 1.63]) were associated with greater ORs for CD/dementia in women than men. CONCLUSIONS Several risk factors were associated with CD/dementia. Higher waist circumference and mental health symptoms were more strongly associated with CD/dementia in women than men. Further studies should examine the mechanisms that underlie these sex differences.
Collapse
Affiliation(s)
- Jessica Gong
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Katie Harris
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Maree Hackett
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
- Faculty of Health and Wellbeing, the University of Central Lancashire, Lancashire, UK
| | - Sanne A E Peters
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
- The George Institute for Global Health, Imperial College London, London, UK
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Henry Brodaty
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
- Dementia Centre for Research Collaboration, University of New South Wales, Sydney, New South Wales, Australia
| | - Mark Cooper
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Pavel Hamet
- Montréal Diabetes Research Centre, Centre Hospitalier de l'Université de Montréal, Quebec, Montreal, Canada
| | - Stephen Harrap
- Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Giuseppe Mancia
- Policlinico di Monza and IRCCS Istituto Auxologico Italiano, University of Milano-Bicocca, Milan, Italy
| | - Stephen MacMahon
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
- The George Institute for Global Health, Imperial College London, London, UK
| | - John Chalmers
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Mark Woodward
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
- The George Institute for Global Health, Imperial College London, London, UK
- Department of Epidemiology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
34
|
Barbiellini Amidei C, Fayosse A, Dumurgier J, Machado-Fragua MD, Tabak AG, van Sloten T, Kivimäki M, Dugravot A, Sabia S, Singh-Manoux A. Association Between Age at Diabetes Onset and Subsequent Risk of Dementia. JAMA 2021; 325:1640-1649. [PMID: 33904867 PMCID: PMC8080220 DOI: 10.1001/jama.2021.4001] [Citation(s) in RCA: 154] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
IMPORTANCE Trends in type 2 diabetes show an increase in prevalence along with younger age of onset. While vascular complications of early-onset type 2 diabetes are known, the associations with dementia remains unclear. OBJECTIVE To determine whether younger age at diabetes onset is more strongly associated with incidence of dementia. DESIGN, SETTING, AND PARTICIPANTS Population-based study in the UK, the Whitehall II prospective cohort study, established in 1985-1988, with clinical examinations in 1991-1993, 1997-1999, 2002-2004, 2007-2009, 2012-2013, and 2015-2016, and linkage to electronic health records until March 2019. The date of final follow-up was March 31, 2019. EXPOSURES Type 2 diabetes, defined as a fasting blood glucose level greater than or equal to 126 mg/dL at clinical examination, physician-diagnosed type 2 diabetes, use of diabetes medication, or hospital record of diabetes between 1985 and 2019. MAIN OUTCOMES AND MEASURES Incident dementia ascertained through linkage to electronic health records. RESULTS Among 10 095 participants (67.3% men; aged 35-55 years in 1985-1988), a total of 1710 cases of diabetes and 639 cases of dementia were recorded over a median follow-up of 31.7 years. Dementia rates per 1000 person-years were 8.9 in participants without diabetes at age 70 years, and rates were 10.0 per 1000 person-years for participants with diabetes onset up to 5 years earlier, 13.0 for 6 to 10 years earlier, and 18.3 for more than 10 years earlier. In multivariable-adjusted analyses, compared with participants without diabetes at age 70, the hazard ratio (HR) of dementia in participants with diabetes onset more than 10 years earlier was 2.12 (95% CI, 1.50-3.00), 1.49 (95% CI, 0.95-2.32) for diabetes onset 6 to 10 years earlier, and 1.11 (95% CI, 0.70-1.76) for diabetes onset 5 years earlier or less; linear trend test (P < .001) indicated a graded association between age at onset of type 2 diabetes and dementia. At age 70, every 5-year younger age at onset of type 2 diabetes was significantly associated with an HR of dementia of 1.24 (95% CI, 1.06-1.46) in analyses adjusted for sociodemographic factors, health behaviors, and health-related measures. CONCLUSIONS AND RELEVANCE In this longitudinal cohort study with a median follow-up of 31.7 years, younger age at onset of diabetes was significantly associated with higher risk of subsequent dementia.
Collapse
Affiliation(s)
- Claudio Barbiellini Amidei
- Epidemiology of Ageing and Neurodegenerative Diseases, Université de Paris, Inserm U1153, Paris, France
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Aurore Fayosse
- Epidemiology of Ageing and Neurodegenerative Diseases, Université de Paris, Inserm U1153, Paris, France
| | - Julien Dumurgier
- Epidemiology of Ageing and Neurodegenerative Diseases, Université de Paris, Inserm U1153, Paris, France
- Cognitive Neurology Center, Lariboisière – Fernand Widal Hospital, AP-HP, Université de Paris, Paris, France
| | - Marcos D. Machado-Fragua
- Epidemiology of Ageing and Neurodegenerative Diseases, Université de Paris, Inserm U1153, Paris, France
| | - Adam G. Tabak
- Department of Epidemiology and Public Health, University College London, United Kingdom
- Department of Internal Medicine and Oncology, Semmelweis University Faculty of Medicine, Budapest, Hungary
- Department of Public Health, Semmelweis University Faculty of Medicine, Budapest, Hungary
| | - Thomas van Sloten
- Cardiovascular Research Institute Maastricht (CARIM), Department of Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Mika Kivimäki
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, Padua, Italy
- Department of Epidemiology and Public Health, University College London, United Kingdom
| | - Aline Dugravot
- Epidemiology of Ageing and Neurodegenerative Diseases, Université de Paris, Inserm U1153, Paris, France
| | - Séverine Sabia
- Epidemiology of Ageing and Neurodegenerative Diseases, Université de Paris, Inserm U1153, Paris, France
- Department of Epidemiology and Public Health, University College London, United Kingdom
| | - Archana Singh-Manoux
- Epidemiology of Ageing and Neurodegenerative Diseases, Université de Paris, Inserm U1153, Paris, France
- Department of Epidemiology and Public Health, University College London, United Kingdom
| |
Collapse
|
35
|
Katsiki N, Kotsa K, Stoian AP, Mikhailidis DP. Hypoglycaemia and Cardiovascular Disease Risk in Patients with Diabetes. Curr Pharm Des 2021; 26:5637-5649. [PMID: 32912117 DOI: 10.2174/1381612826666200909142658] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 08/09/2020] [Indexed: 12/12/2022]
Abstract
Hypoglycaemia represents an important side effect of insulin therapy and insulin secretagogues. It can occur in both type 1 and type 2 diabetes mellitus patients. Also, some associations between hypoglycaemia and cardiovascular (CV) risk have been reported. Several mechanisms may be involved, including the sympathoadrenal system, hypokalaemia, endothelial dysfunction, coagulation, platelets, inflammation, atherothrombosis and impaired autonomic cardiac reflexes. This narrative review discusses the associations of hypoglycaemia with CV diseases, including coronary heart disease (CHD), cardiac arrhythmias, stroke, carotid disease and peripheral artery disease (PAD), as well as with dementia. Severe hypoglycaemia has been related to CHD, CV and all-cause mortality. Furthermore, there is evidence supporting an association between hypoglycaemia and cardiac arrhythmias, potentially predisposing to sudden death. The data linking hypoglycaemia with stroke, carotid disease and PAD is limited. Several factors may affect the hypoglycaemia-CV relationships, such as the definition of hypoglycaemia, patient characteristics, co-morbidities (including chronic kidney disease) and antidiabetic drug therapy. However, the association between hypoglycaemia and dementia is bilateral. Both the disorders are more common in the elderly; thus, glycaemic goals should be carefully selected in older patients. Further research is needed to elucidate the impact of hypoglycaemia on CV disease.
Collapse
Affiliation(s)
- Niki Katsiki
- Division of Endocrinology and Metabolism, Diabetes Center, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Kalliopi Kotsa
- Division of Endocrinology and Metabolism, Diabetes Center, First Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Anca P Stoian
- Diabetes, Nutrition and Metabolic diseases Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom
| |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW The current review evaluates the recent literature on the impact of metabolic dysfunction in human cognition, focusing on epidemiological studies and meta-analyses of these. RECENT FINDINGS Worldwide around 50 million people live with dementia, a number projected to triple by 2050. Recent reports from the Lancet Commission suggest that 40% of dementia cases may be preventable primarily by focusing on well established metabolic dysfunction components and cardiovascular risk factors. SUMMARY There is robust evidence that type 2 diabetes and midlife hypertension increase risk of dementia in late life. Obesity and elevated levels of LDL cholesterol in midlife probably increase risk of dementia, but further research is needed in these areas. Physical activity, diet, alcohol, and smoking might also influence the risk of dementia through their effect on metabolic dysfunction. A key recommendation is to be ambitious about prevention, focusing on interventions to promote healthier lifestyles combating metabolic dysfunction. Only comprehensive multidomain and staff-requiring interventions are however efficient to maintain or improve cognition in at-risk individuals and will be unrealistic economic burdens for most societies to implement. Therefore, a risk score that identifies high-risk individuals will enable a targeted early intensive intervention toward those high-risk individuals that will benefit the most from a prevention against cardiovascular risk factors and metabolic dysfunction.
Collapse
Affiliation(s)
| | | | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Rigshospitalet
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
37
|
Fan D, Yang S, Han Y, Zhang R, Yang L. Isoflurane-induced expression of miR-140-5p aggravates neurotoxicity in diabetic rats by targeting SNX12. J Toxicol Sci 2020; 45:69-76. [PMID: 32062618 DOI: 10.2131/jts.45.69] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
MicroRNAs (miRNAs) are widely known as critical regulators in isoflurane-induced neurotoxicity during the development of brain. Moreover, isoflurane could aggravate cognitive impairment in diabetic rats. The present study was designed to investigate the role and mechanism of miR-140-5p on isoflurane-induced neurotoxicity in diabetic rats. Firstly, a diabetic rat model was established by injection of streptozotocin (STZ) and identified by Morris water maze test. The result indicated that isoflurane treatment exacerbated STZ-induced cognitive impairment, as demonstrated by increase of the latency to the platform and decrease of the proportion of time spent in the target quadrant. Secondly, miR-140-5p was up-regulated in diabetic rats treated with isoflurane. Functional assays revealed that knockdown of miR-140-5p attenuated neurotoxicity in diabetic rats, which was shown by a decrease of the latency to the platform and an increase of the proportion of time spent in the target quadrant. Mechanistically, we demonstrated that miR-140-5p directly bonded to SNX12 (sorting nexin 12). At last, the neuroprotective effect of miR-140-5p knockdown against isoflurane-aggravated neurotoxicity in diabetic rats was dependent on up-regulation of SNX12 and inhibition of cell apoptosis. In summary, these meaningful results demonstrated the mitigation of miR-140-5p knockdown against isoflurane-aggravated neurotoxicity in diabetic rats via SNX12, suggesting a novel target for neuroprotection in diabetes under isoflurane treatment.
Collapse
Affiliation(s)
- Dongyi Fan
- Department of Anesthesiology, the Fifth Affiliated Hospital of Sun Yat-Sen University, China
| | - Simin Yang
- Department of Anesthesiology, the Fifth Affiliated Hospital of Sun Yat-Sen University, China
| | - Yuxiang Han
- Department of Anesthesiology, the Fifth Affiliated Hospital of Sun Yat-Sen University, China
| | - Ru Zhang
- Department of Anesthesiology, the Fifth Affiliated Hospital of Sun Yat-Sen University, China
| | - Lukun Yang
- Department of Anesthesiology, the Fifth Affiliated Hospital of Sun Yat-Sen University, China
| |
Collapse
|
38
|
Giorda CB, Orsi E, De Cosmo S, Bossi AC, Guerzoni C, Cercone S, Gilio B, Cavalot F. Prescription of Sulphonylureas among Patients with Type 2 Diabetes Mellitus in Italy: Results from the Retrospective, Observational Multicentre Cross-Sectional SUSCIPE (Sulphonyl_UreaS_Correct_Internal_Prescription_Evaluation) Study. Diabetes Ther 2020; 11:2105-2119. [PMID: 32734558 PMCID: PMC7434823 DOI: 10.1007/s13300-020-00871-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Indexed: 01/21/2023] Open
Abstract
INTRODUCTION Recent guidelines for the treatment of type 2 diabetes mellitus (T2DM) provide evidence supporting limited use of sulphonylureas (SUs), especially in specific risk patient categories, yet data from national registries still suggest their widespread use. The aim of this study was to investigate characteristics of patients with diabetes treated with SUs and quantify the proportion of patients that met the recommendations for use of SUs by recent guidelines and of those presenting characteristics representing an inappropriate prescription risk (IPR). METHODS A multicenter, retrospective, cross-sectional, observational study in patients with T2DM receiving treatment with SUs (as monotherapy or in combination with another diabetes therapy) was conducted between 2017 and 2018 in 22 outpatient diabetes clinics across Italy. Exclusion criteria were type 1 diabetes, diabetes mellitus secondary to other conditions, and presence of severe/life-threatening diseases. RESULTS A total of 510 patients with T2DM (306 men, 204 women; mean age ± standard deviation 69.8 ± 9.3 years) who were receiving treatment with a SU (as monotherapy or in combination therapy) were assessed in the study. Overall, 70.6% [n = 360; 95% confidence interval (CI) 66.4%, 74.5%] were assessed to have an IPR. Of these, approximately half presented one factor for risk of inappropriate prescription, and 27 and 10.6% presented two and three factors, respectively. In terms of factors contributing to the total burden of risk of inappropriate treatment with SUs, 37.5% (95% CI 33.2%, 41.8%) of all patients were obese; 33.3% (95% CI 29.3%, 37.6%)] were aged ≥ 75 years; 18.6% (95% CI 15.3%, 22.3%) had a history of cardiovascular disease; 14.1% (95% CI 11.2%, 17.4%) had chronic renal insufficiency; 1.8% (95% CI 0.8%, 3.3%) had a history of severe hypoglycemia; 1.8% (95% CI 0.8%; 3.3%) had cognitive impairment; and 2.4% (95% CI 1.2%, 4.1%) had a risky occupation. CONCLUSIONS The results of this study provide evidence of a high rate of inappropriate SU prescription risk among patients with T2DM, especially among those with overweight/obesity, older age, history of cardiovascular disease, and hypoglycemia.
Collapse
Affiliation(s)
| | - Emanuela Orsi
- Department of Medical Science, Endocrinology and Diabetes Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico-University of Milan, Milan, Italy
| | - Salvatore De Cosmo
- Department of Medical Sciences, IRCCS-Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Antonio Carlo Bossi
- Endocrine Diseases Unit-Diabetes Regional Center, Ospedale Treviglio, ASST Bergamo Ovest, Treviglio, BG, Italy
| | | | | | | | - Franco Cavalot
- Metabolic Disease and Diabetes Unit, San Luigi Gonzaga Hospital, Orbassano, TO, Italy
| |
Collapse
|
39
|
Dysfunction of the neurovascular unit in diabetes-related neurodegeneration. Biomed Pharmacother 2020; 131:110656. [PMID: 32841897 DOI: 10.1016/j.biopha.2020.110656] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/10/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022] Open
Abstract
In current aging societies, diabetes mellitus and neurodegenerative diseases represented by Alzheimer's disease are highly prevalent among adults, especially the elderly all over the world. It is worth noting that a substantial body of evidence suggests diabetes contributes to accelerated neurodegenerative processes and the decline of cognition. Over the last few years, some studies have indicated neurovascular uncoupling and disrupted functional connectivity in the early stages of many neurodegenerative diseases, and the concept of the neurovascular unit (NVU) has been highlighted to understand the initiation and progression of neurodegenerative diseases recently. Considering that some components of the NVU are also demonstrated to have abnormal morphology and function under the condition of diabetes, we propose the hypothesis that diabetes may promote the onset and development of neurodegenerative diseases by impairing the integrity of the NVU, named Diabetes-NVU-Neurodegeneration Hypothesis. The existing body of literature supporting the hypothesis and elucidating the underlying mechanisms will be summarized in this review.
Collapse
|
40
|
Livingston G, Huntley J, Sommerlad A, Ames D, Ballard C, Banerjee S, Brayne C, Burns A, Cohen-Mansfield J, Cooper C, Costafreda SG, Dias A, Fox N, Gitlin LN, Howard R, Kales HC, Kivimäki M, Larson EB, Ogunniyi A, Orgeta V, Ritchie K, Rockwood K, Sampson EL, Samus Q, Schneider LS, Selbæk G, Teri L, Mukadam N. Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. Lancet 2020; 396:413-446. [PMID: 32738937 PMCID: PMC7392084 DOI: 10.1016/s0140-6736(20)30367-6] [Citation(s) in RCA: 4866] [Impact Index Per Article: 1216.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 01/31/2020] [Accepted: 02/07/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Gill Livingston
- Division of Psychiatry, University College London, London, UK; Camden and Islington NHS Foundation Trust, London, UK.
| | - Jonathan Huntley
- Division of Psychiatry, University College London, London, UK; Camden and Islington NHS Foundation Trust, London, UK
| | - Andrew Sommerlad
- Division of Psychiatry, University College London, London, UK; Camden and Islington NHS Foundation Trust, London, UK
| | - David Ames
- National Ageing Research Institute and Academic Unit for Psychiatry of Old Age, University of Melbourne, Royal Melbourne Hospital, Parkville, VIC, Australia
| | | | - Sube Banerjee
- Faculty of Health: Medicine, Dentistry and Human Sciences, University of Plymouth, Plymouth, UK
| | - Carol Brayne
- Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Alistair Burns
- Department of Old Age Psychiatry, University of Manchester, Manchester, UK
| | - Jiska Cohen-Mansfield
- Department of Health Promotion, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Heczeg Institute on Aging, Tel Aviv University, Tel Aviv, Israel; Minerva Center for Interdisciplinary Study of End of Life, Tel Aviv University, Tel Aviv, Israel
| | - Claudia Cooper
- Division of Psychiatry, University College London, London, UK; Camden and Islington NHS Foundation Trust, London, UK
| | - Sergi G Costafreda
- Division of Psychiatry, University College London, London, UK; Camden and Islington NHS Foundation Trust, London, UK
| | - Amit Dias
- Department of Preventive and Social Medicine, Goa Medical College, Goa, India
| | - Nick Fox
- Dementia Research Centre, UK Dementia Research Institute, University College London, London, UK; Institute of Neurology, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Laura N Gitlin
- Center for Innovative Care in Aging, Johns Hopkins University, Baltimore, MA, USA
| | - Robert Howard
- Division of Psychiatry, University College London, London, UK; Camden and Islington NHS Foundation Trust, London, UK
| | - Helen C Kales
- Department of Psychiatry and Behavioral Sciences, UC Davis School of Medicine, University of California, Sacramento, CA, USA
| | - Mika Kivimäki
- Department of Epidemiology and Public Health, University College London, London, UK
| | - Eric B Larson
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | | | - Vasiliki Orgeta
- Division of Psychiatry, University College London, London, UK
| | - Karen Ritchie
- Inserm, Unit 1061, Neuropsychiatry: Epidemiological and Clinical Research, La Colombière Hospital, University of Montpellier, Montpellier, France; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Kenneth Rockwood
- Centre for the Health Care of Elderly People, Geriatric Medicine Dalhousie University, Halifax, NS, Canada
| | - Elizabeth L Sampson
- Division of Psychiatry, University College London, London, UK; Barnet, Enfield, and Haringey Mental Health Trust, London, UK
| | - Quincy Samus
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MA, USA
| | - Lon S Schneider
- Department of Psychiatry and the Behavioural Sciences and Department of Neurology, Keck School of Medicine, Leonard Davis School of Gerontology of the University of Southern California, Los Angeles, CA, USA
| | - Geir Selbæk
- Norwegian National Advisory Unit on Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Geriatric Department, Oslo University Hospital, Oslo, Norway
| | - Linda Teri
- Department Psychosocial and Community Health, School of Nursing, University of Washington, Seattle, WA, USA
| | - Naaheed Mukadam
- Division of Psychiatry, University College London, London, UK; Camden and Islington NHS Foundation Trust, London, UK
| |
Collapse
|
41
|
Walking Speed is the Sole Determinant Criterion of Sarcopenia of Mild Cognitive Impairment in Japanese Elderly Patients with Type 2 Diabetes Mellitus. J Clin Med 2020; 9:jcm9072133. [PMID: 32640726 PMCID: PMC7408848 DOI: 10.3390/jcm9072133] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/23/2020] [Accepted: 06/28/2020] [Indexed: 12/19/2022] Open
Abstract
Diabetes mellitus is a risk factor for mild cognitive impairment (MCI) and dementia. However, how the clinical characteristics of MCI patients with type 2 diabetes mellitus are linked to sarcopenia and/or its criteria remain to be elucidated. Japanese patients with type 2 diabetes mellitus were categorized into the MCI group for MoCA-J (the Japanese version of the Montreal cognitive assessment) score <26, and into the non-MCI group for MoCA-J ≥26. Sarcopenia was defined by a low skeletal mass index along with low muscle strength (handgrip strength) or low physical performance (walking speed <1.0 m/s). Univariate and multivariate-adjusted odds ratio models were used to determine the independent contributors for MoCA-J <26. Among 438 participants, 221 (50.5%) and 217 (49.5%) comprised the non-MCI and MCI groups, respectively. In the MCI group, age (61 ± 12 vs. 71 ± 10 years, p < 0.01) and duration of diabetes mellitus (14 ± 9 vs. 17 ± 9 years, p < 0.01) were higher than those in the non-MCI group. Patients in the MCI group exhibited lower hand grip strength, walking speed, and skeletal mass index, but higher prevalence of sarcopenia. Only walking speed (rather than muscle loss or muscle weakness) was found to be an independent determinant of MCI after adjusting for multiple factors, such as age, gender, body mass index (BMI), duration of diabetes mellitus, hypertension, dyslipidemia, smoking, drinking, estimated glomerular filtration rate (eGFR), HbA1c, and history of coronary heart diseases and stroke. In subgroup analysis, a group consisting of male patients aged ≥65 years, with BMI <25, showed a significant OR for walking speed. This study showed that slow walking speed is a sole determinant criterion of sarcopenia of MCI in patients with type 2 diabetes mellitus. It was suggested that walking speed is an important factor in the prediction and prevention of MCI development in patients with diabetes mellitus.
Collapse
|
42
|
Srikanth V, Sinclair AJ, Hill-Briggs F, Moran C, Biessels GJ. Type 2 diabetes and cognitive dysfunction-towards effective management of both comorbidities. Lancet Diabetes Endocrinol 2020; 8:535-545. [PMID: 32445740 DOI: 10.1016/s2213-8587(20)30118-2] [Citation(s) in RCA: 210] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/03/2020] [Accepted: 03/25/2020] [Indexed: 12/25/2022]
Abstract
Type 2 diabetes and cognitive dysfunction are highly prevalent disorders worldwide. Although type 2 diabetes is associated with an increased risk of dementia, awareness of the link between the two conditions is poor, and few recommendations are available to guide clinicians about how to approach cognitive dysfunction in people with diabetes. Clinical guidelines in diabetes have only recently begun to emphasise the importance of cognitive impairment in diabetes and its management. This Series paper aims to synthesise knowledge about the link between diabetes and cognitive dysfunction, issues pertaining to screening and diagnosis of cognitive impairment and dementia in those with type 2 diabetes, management of diabetes in people with cognitive dysfunction (accounting for age and frailty), and emerging therapies for prevention. A conceptual framework for approaching screening and diagnosis is included, and future research directions to guide the field forward are suggested.
Collapse
Affiliation(s)
- Velandai Srikanth
- Department of Geriatric Medicine, Peninsula Health, Peninsula Clinical School, Monash University, Melbourne, VIC, Australia.
| | - Alan J Sinclair
- Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Felicia Hill-Briggs
- Johns Hopkins School of Medicine, Welch Center for Prevention, Epidemiology, and Clinical Research, Baltimore, MD, USA
| | - Chris Moran
- Department of Geriatric Medicine, Peninsula Health, Peninsula Clinical School, Monash University, Melbourne, VIC, Australia
| | - Geert Jan Biessels
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center, Utrecht, Netherlands
| |
Collapse
|
43
|
Sluggett JK, Koponen M, Bell JS, Taipale H, Tanskanen A, Tiihonen J, Uusitupa M, Tolppanen AM, Hartikainen S. Metformin and Risk of Alzheimer's Disease Among Community-Dwelling People With Diabetes: A National Case-Control Study. J Clin Endocrinol Metab 2020; 105:5645285. [PMID: 31778170 DOI: 10.1210/clinem/dgz234] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 11/27/2019] [Indexed: 02/04/2023]
Abstract
CONTEXT Type 2 diabetes has been linked with an increased risk of Alzheimer's disease (AD). Studies on the association between metformin use and AD have reported conflicting results. OBJECTIVE To investigate whether metformin use modifies the association between diabetes and incident, clinically verified AD. DESIGN Nested case-control study. SETTING All community-dwelling people in Finland. PARTICIPANTS Cases were all community-dwelling Finns with AD diagnosed from 2005 to 2011 and with diabetes diagnosed ≥ 3 years before AD (n = 9862). Cases were matched with up to 2 control persons by age, sex, and diabetes duration (n = 19 550). MAIN OUTCOME MEASURE Cumulative metformin exposure was determined from reimbursed dispensings over a 10- to 16-year period. Adjusted odds ratios (aORs) were calculated using conditional logistic regression to estimate associations, with adjustment for potential confounders. RESULTS A total of 7225 (73.3%) cases and 14528 (74.3%) controls received metformin at least once. Metformin use (ever use) was not associated with incident AD (aOR 0.99; 95% confidence interval [CI], 0.94-1.05). The adjusted odds of AD were lower among people dispensed metformin for ≥ 10 years (aOR 0.85; 95% CI, 0.76-0.95), those dispensed cumulative defined daily doses (DDDs) of < 1825-3650 (aOR 0.91; 95% CI, 0.84-0.98) and > 3650 DDDs (aOR 0.77; 95% CI, 0.67-0.88), and among persons dispensed an average of 2 g metformin daily (aOR 0.89; 95% CI, 0.82-0.96). CONCLUSION In this large national sample we found no evidence that metformin use increases the risk of AD. Conversely, long-term and high-dose metformin use was associated with a lower risk of incident AD in older people with diabetes.
Collapse
Affiliation(s)
- Janet K Sluggett
- Centre for Medicine Use and Safety, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- NHMRC Cognitive Decline Partnership Centre, Hornsby Ku-ring-gai Hospital, Hornsby, New South Wales, Australia
| | - Marjaana Koponen
- Centre for Medicine Use and Safety, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Kuopio Research Centre for Geriatric Care, University of Eastern Finland, Kuopio, Finland
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - J Simon Bell
- Centre for Medicine Use and Safety, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- NHMRC Cognitive Decline Partnership Centre, Hornsby Ku-ring-gai Hospital, Hornsby, New South Wales, Australia
- Kuopio Research Centre for Geriatric Care, University of Eastern Finland, Kuopio, Finland
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Heidi Taipale
- Kuopio Research Centre for Geriatric Care, University of Eastern Finland, Kuopio, Finland
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Antti Tanskanen
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Public Health Evaluation and Projection, National Institute for Health and Welfare, Helsinki, Finland
| | - Jari Tiihonen
- Department of Forensic Psychiatry, University of Eastern Finland, Niuvanniemi Hospital, Kuopio, Finland
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| | - Matti Uusitupa
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Anna-Maija Tolppanen
- Kuopio Research Centre for Geriatric Care, University of Eastern Finland, Kuopio, Finland
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Sirpa Hartikainen
- Kuopio Research Centre for Geriatric Care, University of Eastern Finland, Kuopio, Finland
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
44
|
Abdelhafiz AH, Davies PC, Sinclair AJ. Triad of impairment in older people with diabetes-reciprocal relations and clinical implications. Diabetes Res Clin Pract 2020; 161:108065. [PMID: 32044347 DOI: 10.1016/j.diabres.2020.108065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 01/28/2020] [Accepted: 02/06/2020] [Indexed: 12/25/2022]
Abstract
Frailty is emerging as a new category complication of diabetes in older people. Clinically, frailty is still not well defined and mostly viewed as a decline in solely the physical domain. However, frailty is a multidimensional syndrome and the newly introduced concept of "triad of impairment" (physical, cognitive and emotional) may be a more representative of the broad nature of frailty. The components of the triad of impairment (TOI) commonly coexist and demonstrate a reciprocal relation. Diabetes in old age appears to increase the risk of the triad of impairment, which may eventually progress to disability. Therefore, older people with diabetes should be regularly assessed for the presence of these three key components. Adequate nutrition and regular resistance exercise training have been shown to have a positive impact on the long-term outcome in this population. However, the role of good glycaemic control and the use of current hypoglycaemic medications in reducing the incidence of this triad are less clear. Future research is needed to develop novel hypoglycaemic medications that not only focus on glycaemic control and cardiovascular safety but also on reducing the risk of the triad of impairment.
Collapse
Affiliation(s)
- A H Abdelhafiz
- Department of Geriatric Medicine, Rotherham General Hospital, Moorgate Road, Rotherham S60 2UD, UK.
| | - P C Davies
- Department of Geriatric Medicine, Rotherham General Hospital, Moorgate Road, Rotherham S60 2UD, UK
| | - A J Sinclair
- Foundation for Diabetes Research in Older People, Diabetes Frail Ltd, Droitwich Spa WR9 0QH, UK; Kings College, London SE1 9NH, UK
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Dementia is rapidly growing as sources of morbidity and mortality as the US population ages, but its pathophysiology remains poorly understood. As a result, no disease-modifying treatments currently exist. We review the evidence that nonesterified fatty acids may play a key role in this condition. RECENT FINDINGS Nonesterified fatty acids appear to influence several pathways leading to dementia. In addition to their vascular effects, these moieties cross the blood-brain barrier, where they are toxic to several cell types. They may also influence insulin metabolism in the brain directly and indirectly, and some drugs that lower circulating levels appear to slow cognitive decline and brain atrophy in diabetes. SUMMARY Nonesterified fatty acids may contribute to dementia, much as they do to diabetes and cardiovascular disease. Several therapeutic agents lower circulating levels of nonesterified fatty acids and should be tested for their potential preventive effects on cognitive decline in healthy populations before irreversible neuronal attrition occurs.
Collapse
Affiliation(s)
- Kenneth J Mukamal
- Beth Israel Deaconess Medical Center, General Medicine, Brookline, Massachusetts, USA
| |
Collapse
|
46
|
Abstract
Cognitive dysfunction, including mild cognitive impairment and dementia, is increasingly recognised as an important comorbidity and complication of diabetes that affects an individual's well-being and diabetes management, and is associated with diabetes treatment-related complications. Recent guidelines therefore recommend screening for cognitive impairment in older individuals with diabetes. In addition, these guidelines suggest that glucose-lowering treatment should be tailored in those diagnosed with cognitive impairment, to reduce the risk of hypoglycaemia and improve treatment adherence. This review gives an overview of cognitive dysfunction in people with diabetes, briefly describing the clinical features of different stages of cognitive dysfunction and their epidemiology. In particular, it addresses essential additional steps that need to be taken to fully implement the emerging guidelines on screening and management of cognitive dysfunction in diabetes into daily practice.
Collapse
Affiliation(s)
- Geert J Biessels
- Department of Neurology, G03.232, UMC Utrecht Brain Center, University Medical Center Utrecht, PO Box 85500, 3508 GA, Utrecht, the Netherlands.
| | - Rachel A Whitmer
- Department of Public Health Sciences, Division of Epidemiology, Population Brain Health Laboratory, University of California Davis, Davis, CA, USA
| |
Collapse
|
47
|
Rorbach-Dolata A, Piwowar A. Neurometabolic Evidence Supporting the Hypothesis of Increased Incidence of Type 3 Diabetes Mellitus in the 21st Century. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1435276. [PMID: 31428627 PMCID: PMC6679855 DOI: 10.1155/2019/1435276] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 07/03/2019] [Indexed: 12/11/2022]
Abstract
The most recent evidence supports the existence of a link between type 2 diabetes (T2DM) and Alzheimer's Disease (AD), described by the new term: type 3 diabetes (T3D). The increasing incidence of T2DM in the 21st century and accompanying reports on the higher risk of AD in diabetic patients prompts the search for pathways linking glycemia disturbances and neurodegeneration. It is suggested that hyperglycemia may lead to glutamate-induced excitotoxicity, a pathological process resulting from excessive depolarization of membrane and uncontrolled calcium ion influx into neuronal cells. On the other hand, it has been confirmed that peripheral insulin resistance triggers insulin resistance in the brain, which may consequently contribute to AD by amyloid beta accumulation, tau phosphorylation, oxidative stress, advanced glycation end products, and apoptosis. Some literature sources suggest significant amylin involvement in additional amyloid formation in the central nervous system, especially under hyperamylinemic conditions. It is particularly important to provide early diagnostics in people with metabolic disturbances, especially including fasting insulin and HOMA-IR, which are necessary to reveal insulin resistance. The present review reveals the most recent and important evidence associated with the phenomenon of T3D and discusses the potential lacks of prevention and diagnostics for diabetes which might result in neurometabolic disorders, from a pharmacotherapy perspective.
Collapse
Affiliation(s)
- Anna Rorbach-Dolata
- Department of Toxicology, Faculty of Pharmacy with the Division of Laboratory Diagnostics, Wroclaw Medical University, Borowska 211, 50-552 Wroclaw, Poland
| | - Agnieszka Piwowar
- Department of Toxicology, Faculty of Pharmacy with the Division of Laboratory Diagnostics, Wroclaw Medical University, Borowska 211, 50-552 Wroclaw, Poland
| |
Collapse
|