1
|
van Gils V, Tao Q, Ang TF, Young CB, Mormino EC, Qiu WQ, Visser PJ, Au R, Jansen WJ, Vos SJ. Associations Between Glucose Metabolism Measures and Amyloid-β and Tau Load on PET 14 Years Later: Findings From the Framingham Heart Study. Diabetes Care 2024; 47:1787-1793. [PMID: 39078159 PMCID: PMC11417279 DOI: 10.2337/dc24-0162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/03/2024] [Indexed: 07/31/2024]
Abstract
OBJECTIVE Type 2 diabetes and glucose metabolism have previously been linked to Alzheimer disease (AD). Yet, findings on the relation of glucose metabolism with amyloid-β and tau pathology later in life remain unclear. RESEARCH DESIGN AND METHODS We included 288 participants (mean age 43.1 years, SD 10.7, range 20-70 years) without dementia, from the Framingham Heart Study, who had available measures of glucose metabolism (i.e., one-time fasting plasma glucose and insulin) and positron emission tomography (PET) measures of amyloid-β and/or tau 14 years later. We performed linear regression analyses to test associations of plasma glucose (continuously and categorically; elevated defined as >100 mg/dL), plasma insulin, homeostatic model assessment for insulin resistance (HOMA-IR) with amyloid-β or tau load on PET. When significant, we explored whether age, sex, and APOE ε4 allele carriership (AD genetic risk) modified these associations. RESULTS Our findings indicated that elevated plasma glucose was associated with greater tau load 14 years later (B [95% CI] = 0.03 [0.01-0.05], P = 0.024 after false discovery rate [FDR] correction) but not amyloid-β. APOE ε4 carriership modified this association (B [95% CI] = -0.08 [-0.12 to -0.03], P = 0.001), indicating that the association was only present in APOE ε4 noncarriers (n = 225). Plasma insulin and HOMA-IR were not associated with amyloid-β or tau load 14 years later after FDR correction. CONCLUSIONS Our findings suggest that glucose metabolism is associated with increased future tau but not amyloid-β load. This provides relevant knowledge for prevention strategies and prognostics to improve health care.
Collapse
Affiliation(s)
- Veerle van Gils
- Alzheimer Center Limburg, School for Mental Health and Neuroscience, Department of Psychiatry & Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Qiushan Tao
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
- Slone Epidemiology Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
| | - Ting F.A. Ang
- Slone Epidemiology Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
| | - Christina B. Young
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA
| | - Elizabeth C. Mormino
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA
| | - Wei Qiao Qiu
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
| | - Pieter Jelle Visser
- Alzheimer Center Limburg, School for Mental Health and Neuroscience, Department of Psychiatry & Neuroscience, Maastricht University, Maastricht, the Netherlands
- Alzheimer Center and Department of Neurology, Amsterdam Neuroscience Campus, VU University Medical Center, Amsterdam, the Netherlands
| | - Rhoda Au
- Slone Epidemiology Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
- Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
- Department of Neurology & Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA
| | - Willemijn J. Jansen
- Alzheimer Center Limburg, School for Mental Health and Neuroscience, Department of Psychiatry & Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Stephanie J.B. Vos
- Alzheimer Center Limburg, School for Mental Health and Neuroscience, Department of Psychiatry & Neuroscience, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
2
|
van Gils V, Rizzo M, Côté J, Viechtbauer W, Fanelli G, Salas-Salvadó J, Wimberley T, Bulló M, Fernandez-Aranda F, Dalsgaard S, Visser PJ, Jansen WJ, Vos SJB. The association of glucose metabolism measures and diabetes status with Alzheimer's disease biomarkers of amyloid and tau: A systematic review and meta-analysis. Neurosci Biobehav Rev 2024; 159:105604. [PMID: 38423195 DOI: 10.1016/j.neubiorev.2024.105604] [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: 11/23/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/02/2024]
Abstract
Conflicting evidence exists on the relationship between diabetes mellitus (DM) and Alzheimer's disease (AD) biomarkers. Therefore, we conducted a random-effects meta-analysis to evaluate the correlation of glucose metabolism measures (glycated hemoglobin, fasting blood glucose, insulin resistance indices) and DM status with AD biomarkers of amyloid-β and tau measured by positron emission tomography or cerebrospinal fluid. We selected 37 studies from PubMed and Embase, including 11,694 individuals. More impaired glucose metabolism and DM status were associated with higher tau biomarkers (r=0.11[0.03-0.18], p=0.008; I2=68%), but were not associated with amyloid-β biomarkers (r=-0.06[-0.13-0.01], p=0.08; I2=81%). Meta-regression revealed that glucose metabolism and DM were specifically associated with tau biomarkers in population settings (p=0.001). Furthermore, more impaired glucose metabolism and DM status were associated with lower amyloid-β biomarkers in memory clinic settings (p=0.004), and in studies with a higher prevalence of dementia (p<0.001) or lower cognitive scores (p=0.04). These findings indicate that DM is associated with biomarkers of tau but not with amyloid-β. This knowledge is valuable for improving dementia and DM diagnostics and treatment.
Collapse
Affiliation(s)
- Veerle van Gils
- Department of Psychiatry & Neuropsychology, Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.
| | - Marianna Rizzo
- Department of Psychiatry & Neuropsychology, Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Jade Côté
- Department of Psychiatry & Neuropsychology, Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Wolfgang Viechtbauer
- Department of Psychiatry & Neuropsychology, Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Giuseppe Fanelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy; Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Nijmegen, the Netherlands
| | - Jordi Salas-Salvadó
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Alimentació, Nutrició, Desenvolupament i Salut Mental (ANUT-DSM), Unitat de Nutrició Humana, Reus, Spain; Institut d'Investigació Sanitària Pere Virgili (IISPV), Alimentació, Nutrició, Desenvolupament i Salut Mental, Reus, Spain; CIBER Physiology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, Madrid 28029, Spain
| | - Theresa Wimberley
- The National Center for Register-based Research, School of Business and Social Sciences, Aarhus University, Aarhus, Denmark
| | - Mònica Bulló
- CIBER Physiology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, Madrid 28029, Spain; Nutrition and Metabolic Health Research Group (NuMeH). Department of Biochemistry and Biotechnology, Rovira i Virgili University (URV), Reus 43201, Spain; Center of Environmental, Food and Toxicological Technology - TecnATox, Rovira i Virgili University, Reus 43201, Spain
| | - Fernando Fernandez-Aranda
- CIBER Physiology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, Madrid 28029, Spain; Department of Clinical Psychology, Bellvitge University Hospital-IDIBELL, Barcelona, Spain; Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Spain
| | - Søren Dalsgaard
- The National Center for Register-based Research, School of Business and Social Sciences, Aarhus University, Aarhus, Denmark; iPSYCH - The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark
| | - Pieter Jelle Visser
- Department of Psychiatry & Neuropsychology, Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands; Alzheimer Center and Department of Neurology, Amsterdam Neuroscience Campus, VU University Medical Center, Amsterdam, the Netherlands
| | - Willemijn J Jansen
- Department of Psychiatry & Neuropsychology, Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Stephanie J B Vos
- Department of Psychiatry & Neuropsychology, Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
3
|
Zou J, Niu K, Lu T, Kan J, Cheng H, Xu L. The Multifunction of TRIM26: From Immune Regulation to Oncology. Protein Pept Lett 2024; 31:424-436. [PMID: 38956921 PMCID: PMC11475100 DOI: 10.2174/0109298665311516240621114519] [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: 02/20/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 07/04/2024]
Abstract
Ubiquitination, a crucial post-translational modification, plays a role in nearly all physiological processes. Its functional execution depends on a series of catalytic reactions involving numerous proteases. TRIM26, a protein belonging to the TRIM family, exhibits E3 ubiquitin ligase activity because of its RING structural domain, and is present in diverse cell lineages. Over the last few decades, TRIM26 has been documented to engage in numerous physiological and pathological processes as a controller, demonstrating a diverse array of biological roles. Despite the growing research interest in TRIM26, there has been limited attention given to examining the protein's structure and function in existing reviews. This review begins with a concise overview of the composition and positioning of TRIM26 and then proceeds to examine its roles in immune response, viral invasion, and inflammatory processes. Simultaneously, we demonstrate the contribution of TRIM26 to the progression of various diseases, encompassing numerous malignancies and neurologic conditions. Finally, we have investigated the potential areas for future research on TRIM26.
Collapse
Affiliation(s)
- Jialai Zou
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Kaiyi Niu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Tao Lu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Jianxun Kan
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Hao Cheng
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| | - Lijian Xu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
| |
Collapse
|
4
|
Chen YH, Wang ZB, Liu XP, Mao ZQ. Plasma Insulin Predicts Early Amyloid-β Pathology Changes in Alzheimer's Disease. J Alzheimers Dis 2024; 100:321-332. [PMID: 38848190 DOI: 10.3233/jad-240289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
Background Evidence suggests that type 2 diabetes (T2D) is an independent risk factor for Alzheimer's disease (AD), sharing similar pathophysiological traits like impaired insulin signaling. Objective To test the association between plasma insulin and cerebrospinal fluid (CSF) AD pathology. Methods A total of 304 participants were included in the Alzheimer's Disease Neuroimaging Initiative, assessing plasma insulin and CSF AD pathology. We explored the cross-sectional and longitudinal associations between plasma insulin and AD pathology and compared their associations across different AD clinical and pathological stages. Results In the non-demented group, amyloid-β (Aβ)+ participants (e.g., as reflected by CSF Aβ42) exhibited significantly lower plasma insulin levels compared to non-demented Aβ-participants (p < 0.001). This reduction in plasma insulin was more evident in the A+T+ group (as shown by CSF Aβ42 and pTau181 levels) when compared to the A-T- group within the non-dementia group (p = 0.002). Additionally, higher plasma insulin levels were consistently associated with more normal CSF Aβ42 levels (p < 0.001) across all participants. This association was particularly significant in the Aβ-group (p = 0.002) and among non-demented individuals (p < 0.001). Notably, baseline plasma insulin was significantly correlated with longitudinal changes in CSF Aβ42 (p = 0.006), whereas baseline CSF Aβ42 did not show a similar correlation with changes in plasma insulin over time. Conclusions These findings suggest an association between plasma insulin and early Aβ pathology in the early stages of AD, indicating that plasma insulin may be a potential predictor of changes in early Aβ pathology.
Collapse
Affiliation(s)
- Yu-Han Chen
- The First Clinical Medical School, Hebei North University, Zhangjiakou, China
| | - Zhi-Bo Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Xi-Peng Liu
- Department of Neurosurgery, The First Affiliated Hospital of Hebei North University, Hebei, Zhangjiakou, China
| | - Zhi-Qi Mao
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
5
|
Pietilä E, Snellman A, Tuisku J, Helin S, Viitanen M, Jula A, Rinne JO, Ekblad LL. Midlife insulin resistance, APOE genotype, and change in late-life brain beta-amyloid accumulation - A 5-year follow-up [ 11C]PIB-PET study. Neurobiol Dis 2024; 190:106385. [PMID: 38123104 DOI: 10.1016/j.nbd.2023.106385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/14/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
We studied if midlife insulin resistance (IR) and APOE genotype would predict brain beta-amyloid (Aβ) accumulation and Aβ change in late-life in 5-year follow-up [11C]PIB-PET study. 43 dementia-free participants were scanned twice with [11C]PIB-PET in their late-life (mean age at follow-up 75.4 years). Participants were recruited from the Finnish Health2000 study according to their HOMA-IR values measured in midlife (mean age at midlife 55.4 years; IR+ group, HOMA-IR > 2.17; IR- group, HOMA-IR <1.25), and their APOEε4 genotype. At late-life follow-up, [11C]PIB-PET composite SUVr was significantly higher in IR+ group than IR- group (median 2.3 (interquartile range 1.7-3.3) vs. 1.7 (1.5-2.4), p = 0.03). There was no difference between IR- and IR+ groups in [11C]PIB-PET SUVr 5-year change, but the change was significantly higher in IR+/APOEε4+ group (median change 0.8 (0.60-1.0)) than in IR-/APOEε4- (0.28 (0.14-0.47), p = 0.02) and in IR+/APOEε4- group (0.24 (0.06-0.40), p = 0.046). These results suggest that APOEε4 carriers with midlife IR are at increased risk for late-life Aβ accumulation.
Collapse
Affiliation(s)
- Elina Pietilä
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland.
| | - Anniina Snellman
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Jouni Tuisku
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Semi Helin
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Matti Viitanen
- Department of Geriatrics, Turku City Hospital and University of Turku, Finland; Division of Clinical Geriatrics, NVS, Karolinska Institutet, Stockholm, Sweden
| | - Antti Jula
- Finnish Institute for Health and Welfare, Turku, Finland
| | - Juha O Rinne
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland; InFLAMES Reseach Flagship Center, University of Turku, Turku, Finland
| | - Laura L Ekblad
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland; Department of Geriatrics, Turku University Hospital, Wellbeing services county of Southwestern Finland, Finland
| |
Collapse
|
6
|
Norgren J, Sindi S, Matton A, Kivipelto M, Kåreholt I. APOE-Genotype and Insulin Modulate Estimated Effect of Dietary Macronutrients on Cognitive Performance: Panel Analyses in Nondiabetic Older Adults at Risk of Dementia. J Nutr 2023; 153:3506-3520. [PMID: 37778510 DOI: 10.1016/j.tjnut.2023.09.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/20/2023] [Accepted: 09/07/2023] [Indexed: 10/03/2023] Open
Abstract
BACKGROUND The apolipoprotein E gene (APOE ε-2/3/4, combined as 6 different genotypes: ε-22/23/24/33/34/44) and insulin status modulate dementia risk and play a role in the metabolism of macronutrients. OBJECTIVES We aimed to examine APOE-genotype and fasting insulin as effect modifiers of the slopes between dietary macronutrients and cognitive performance among older adults at risk of dementia. METHODS Panel analyses-with diet and cognition measured at baseline and follow-up at years 1 and 2-were performed in a sub-sample from the FINGER (Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability) trial (n = 676, 60-77 y, 46% females, all nondiabetics). The associations between macronutrients (3-d food records, z-scores) and global cognition (modified Neuropsychological Test Battery, z-score) were analyzed in mixed regression models adjusted for confounders selected a priori. After a gradient was implied by the point estimates in categorical APOE analyses, we investigated a continuous APOE variable [APOE-gradient, coded -1 (for ε-23), -0.5 (ε-24), 0 (ε-33), 1 (ε-34), 2 (ε-44)] as an effect-modifier. RESULTS At increasing levels of the APOE-gradient, a relatively more favorable slope between diet and cognition was observed for a lower carbohydrate/fat ratio [β = -0.040, 95% confidence interval (CI): -0.074, -0.006; P = 0.020 for interaction diet × APOE-gradient), and higher protein (β = 0.075, 95% CI: 0.042, 0.109; P = 9.4 × 10-6). Insulin concentration (log-linear) modulated the association between the carbohydrate/fat ratio and cognition by a quadratic interaction (β = -0.016, P = 0.039). Coherent findings for exploratory predictors (fiber, fat subtypes, composite score, metabolic biomarkers) were compatible with published hypotheses of differential dietary adaptation by APOE, with cognition among ε-33 being relatively independent of dietary parameters-implying "metabolic flexibility." Antagonistic slopes to cognition for ε-23 (positive) compared with ε-34 and ε-44 (negative) were found for a Higher-carbohydrates-fiber-Lower-fat-protein composite score, even as within-subjects effects. CONCLUSIONS APOE-based precision nutrition appears conceptually promising, but replications in wider samples are warranted, as well as support from trials. Both relative hyper- and hypoinsulinemia might modulate the effect of diet on cognition.
Collapse
Affiliation(s)
- Jakob Norgren
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.
| | - Shireen Sindi
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden; Neuroepidemiology and Ageing Research Unit, School of Public Health, Imperial College London, London, United Kingdom
| | - Anna Matton
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden; Neuroepidemiology and Ageing Research Unit, School of Public Health, Imperial College London, London, United Kingdom; Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Miia Kivipelto
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden; Neuroepidemiology and Ageing Research Unit, School of Public Health, Imperial College London, London, United Kingdom; Theme Inflammation and Aging, Medical Unit Aging, Karolinska University Hospital, Stockholm, Sweden; Stockholms Sjukhem, Research and Development Unit, Stockholm, Sweden
| | - Ingemar Kåreholt
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden; Institute of Gerontology, School of Health and Welfare, Jönköping University, Jönköping, Sweden; Department of Neurobiology, Care Sciences and Society, Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| |
Collapse
|
7
|
Eruysal E, Ravdin L, Zhang C, Kamel H, Iadecola C, Ishii M. Sexually Dimorphic Association of Circulating Plasminogen Activator Inhibitor-1 Levels and Body Mass Index with Cerebrospinal Fluid Biomarkers of Alzheimer's Pathology in Preclinical Alzheimer's Disease. J Alzheimers Dis 2023; 91:1073-1083. [PMID: 36565112 PMCID: PMC10518184 DOI: 10.3233/jad-220686] [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] [Indexed: 12/24/2022]
Abstract
BACKGROUND Plasminogen activator inhibitor-1 (PAI-1), an inhibitor of fibrinolysis that is associated with adiposity, has been implicated in Alzheimer's disease (AD) pathogenesis. However, whether circulating PAI-1 levels are altered during preclinical AD remains unclear. OBJECTIVE To measure plasma PAI-1 levels in cognitively normal cerebrospinal fluid (CSF) AD biomarker positive and biomarker negative participants and to examine the association of plasma PAI-1 levels with CSF AD biomarkers and Mini-Mental State Examination (MMSE) scores. METHODS In this cross-sectional study, plasma PAI-1 levels were measured in 155 cognitively normal (Clinical Dementia Rating, CDR 0) non-obese older adults. 29 men and 26 women were classified as preclinical AD by previously established CSF tau/Aβ42 criteria. All analyses were sex stratified due to reported sex differences in PAI-1 expression. RESULTS Plasma PAI-1 levels were associated with body mass index (BMI) but not age in men and women. In men, plasma PAI-1 levels and BMI were lower in preclinical AD compared to control. Plasma PAI-1 levels were positively associated with CSF amyloid-β42 (Aβ42) and CSF Aβ42/Aβ40 and negatively associated with CSF tau/Aβ42, while BMI was positively associated with CSF Aβ42 and negatively associated with CSF p-tau181 and CSF tau/Aβ42. In women, plasma PAI-1 levels and BMI were similar between preclinical AD and control and were not associated with CSF AD biomarkers. For men and women, plasma PAI-1 levels and BMI were not associated with MMSE scores. CONCLUSION These findings suggest that there are significant sex differences in the systemic metabolic changes seen in the preclinical stage of AD.
Collapse
Affiliation(s)
- Emily Eruysal
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Lisa Ravdin
- Department of Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Cenai Zhang
- Department of Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Hooman Kamel
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Neurology, Weill Cornell Medicine, New York, NY, USA
| | - Makoto Ishii
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Department of Neurology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
8
|
Gao H, Zhou Y, Jin PS, Wu DG, Wang YN, Zhao X, Zhao B. Molecular alteration of the proteasome contributes to AD-like pathology in the brain of HFD-STZ diabetic rats. Metab Brain Dis 2022; 38:1013-1024. [PMID: 36580191 DOI: 10.1007/s11011-022-01151-w] [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: 06/12/2022] [Accepted: 12/21/2022] [Indexed: 12/30/2022]
Abstract
Diabetes-related cognitive impairment has been shown in diverse epidemiological investigations and lab-based studies, although the underlying pathological mechanisms remain unclear. Unbalanced protein homeostasis may contribute to cognitive decline by inducing abnormal protein aggregation in the diabetic brain. This study aimed to determine possible changes in the proteasome, which is an important pathway involved in abnormal protein degradation. To this end, we examined potential alterations of proteasomal subunits and hydrolytic activity in the brain of diabetic rats fed with high-fat diet combined with small doses of streptozotocin (STZ). Furthermore, lactacystin were used to inhibit proteasomal activity in vivo and typical Alzheimer's disease (AD)-like pathologies were detected, including amyloid-beta, tau phosphorylation, and oxidative protein changes. Our results showed that proteasomal activity increased in the brains of diabetic rats compared to age-matched control rats. After proteasome inhibition, the levels of tau phosphorylation and protein oxidative modification significantly increased; however, no changes were detected in the pathway involved in amyloid production. These results indicated that changes in protein homeostasis balance in diabetes play a role in some typical AD-like changes, especially in oxidative protein degradation, providing evidence that prevention of diabetes-induced protein imbalance may be a potential therapeutic target.
Collapse
Affiliation(s)
- Han Gao
- School of Basic Medicine Sciences, Dali University, 6Th Xue-Ren Road, Dali, 671000, Yunnan Province, People's Republic of China
| | - Ye Zhou
- School of Basic Medicine Sciences, Dali University, 6Th Xue-Ren Road, Dali, 671000, Yunnan Province, People's Republic of China
| | - Peng-Shuai Jin
- School of Basic Medicine Sciences, Dali University, 6Th Xue-Ren Road, Dali, 671000, Yunnan Province, People's Republic of China
- Zhalantun Vocational College, 20Th Zhongyang Road, Hulunbuir, NeiMonggol Autonomous Region, People's Republic of China
| | - Dong-Gui Wu
- School of Basic Medicine Sciences, Dali University, 6Th Xue-Ren Road, Dali, 671000, Yunnan Province, People's Republic of China
- Zhuhai City People's Hospital, Zhuhai, Guangdong Province, People's Republic of China
| | - Yu-Na Wang
- School of Basic Medicine Sciences, Dali University, 6Th Xue-Ren Road, Dali, 671000, Yunnan Province, People's Republic of China
| | - Xi Zhao
- School of Basic Medicine Sciences, Dali University, 6Th Xue-Ren Road, Dali, 671000, Yunnan Province, People's Republic of China
| | - Bei Zhao
- School of Basic Medicine Sciences, Dali University, 6Th Xue-Ren Road, Dali, 671000, Yunnan Province, People's Republic of China.
- Li Yunqing Expert Workstation of Yunnan Province (No.202005AF150014), Dali University, 6Th Xue-Ren Road, Dali, Yunnan Province, People's Republic of China.
| |
Collapse
|
9
|
Li S, Deng X, Zhang Y. The Triglyceride-Glucose Index Is Associated with Longitudinal Cognitive Decline in a Middle-Aged to Elderly Population: A Cohort Study. J Clin Med 2022; 11:jcm11237153. [PMID: 36498726 PMCID: PMC9737091 DOI: 10.3390/jcm11237153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/24/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND To examine the effect of the triglyceride-glucose (TyG) index on longitudinal cognitive decline in a healthy middle-aged-to-elderly population. METHODS We conducted a population-based longitudinal study. A total of 1774 participants without cognitive impairment were enrolled in the 4-year follow-up. They were divided into four groups according to the quartile of the TyG index. Multivariable-adjusted Cox proportional hazard models were performed to examine the association between the TyG index and cognitive decline. Discrimination tests were used to evaluate the incremental predictive value of the TyG index beyond conventional risk factors. RESULTS During the follow-up, compared with those in the bottom quartile group, participants in the top TyG quartile group presented a 51% increase in the risk of cognitive decline (OR 1.51 (95% CI: 1.06-2.14)). As shown by discrimination tests, adding the TyG index into the conventional model resulted in a slight improvement in predicting the risk of cognitive decline (NRI 16.00% (p = 0.004)). CONCLUSION This study demonstrated that increasing values of the TyG index were positively associated with the risk of cognitive decline. Monitoring the TyG index may help in the early identification of individuals at high risk of cognitive deterioration.
Collapse
Affiliation(s)
- Siqi Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Xuan Deng
- Clinical Research Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yumei Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Department of Rehabilitation Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
- Correspondence: ; Tel.: +86-10-59975531
| |
Collapse
|
10
|
Bi Q, Zhou X, Lu Y, Fu W, Wang Y, Wang F, Wang J. Polymorphisms of the apolipoprotein E gene affect response to atorvastatin therapy in acute ischemic stroke. Front Cardiovasc Med 2022; 9:1024014. [PMID: 36426228 PMCID: PMC9678920 DOI: 10.3389/fcvm.2022.1024014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 10/20/2022] [Indexed: 10/13/2023] Open
Abstract
Background Polymorphisms of the apolipoprotein E (APOE) gene are related to the efficacy of statin therapy. The biological functions of the APOE subtypes determine the metabolism of blood plasma lipids and the progression of atherosclerosis. This study aimed to explore the impact of APOE gene polymorphisms on the effect of atorvastatin on lipid regulation and plaque stabilization. Methods The study was a prospective cohort study that consecutively included patients with acute ischemic stroke (AIS) in the Department of Neurology, Shanghai Tenth People's Hospital, from December 2018 to December 2019. The patients were divided into E2, E3, and E4 groups according to their APOE genotype. Atorvastatin (20 mg) was administrated to all patients. Changes in blood lipid levels over 3 months and plaque size and stability over 12 months were analyzed. Results We enrolled 253 consecutive patients with AIS, of whom, 136 had carotid atherosclerotic plaques. Two patients with genotype E2/E4 were excluded. There were 30 patients in the E2 group (12.0%), 191 patients in the E3 group (76.0%), and 30 patients in the E4 group (12.0%). The lowest percentage reduction in low-density lipoprotein cholesterol (LDL-C) was observed in the E4 group (41.2%), while the highest percentage reduction was observed in the E2 group (17.6%). The plaques in the E2 group showed slower progression, while those in the E4 group showed more rapid progression. Conclusion APOE gene polymorphisms affect the biological functions of atorvastatin. Compared to the ε3 or ε4 allele, the ε2 allele exerted a greater lipid-lowering effect on LDL-C levels, enhanced the ability of atorvastatin to stabilize carotid artery plaques, and slowed carotid artery plaque progression.
Collapse
Affiliation(s)
- QianQian Bi
- Department of Neurology, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - XiaoYu Zhou
- Department of Neurology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - YanQin Lu
- Department of Infectious Diseases, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wang Fu
- Department of Neurology, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - YongPeng Wang
- Department of Neurology, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Feng Wang
- Department of Neurology, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jue Wang
- Department of Neurology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Savelieff MG, Chen KS, Elzinga SE, Feldman EL. Diabetes and dementia: Clinical perspective, innovation, knowledge gaps. J Diabetes Complications 2022; 36:108333. [PMID: 36240668 PMCID: PMC10076101 DOI: 10.1016/j.jdiacomp.2022.108333] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/30/2022] [Indexed: 10/31/2022]
Abstract
The world faces a pandemic-level prevalence of type 2 diabetes. In parallel with this massive burden of metabolic disease is the growing prevalence of dementia as the population ages. The two health issues are intertwined. The Lancet Commission on dementia prevention, intervention, and care was convened to tackle the growing global concern of dementia by identifying risk factors. It concluded, along with other studies, that diabetes as well as obesity and the metabolic syndrome more broadly, which are frequently comorbid, raise the risk of developing dementia. Type 2 diabetes is a modifiable risk factor; however, it is uncertain whether anti-diabetic drugs mitigate risk of developing dementia. Reasons are manifold but constitute a critical knowledge gap in the field. This review outlines studies of type 2 diabetes on risk of dementia, illustrating key concepts. Moreover, it identifies knowledge gaps, reviews strategies to help fill these gaps, and concludes with a series of recommendations to mitigate risk and advance understanding of type 2 diabetes and dementia.
Collapse
Affiliation(s)
- Masha G Savelieff
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kevin S Chen
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA; Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Sarah E Elzinga
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA; Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Eva L Feldman
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA; Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
12
|
Kumar P, Mishra J, Kumar N. Mechanistic Role of Jak3 in Obesity-Associated Cognitive Impairments. Nutrients 2022; 14:nu14183715. [PMID: 36145091 PMCID: PMC9505565 DOI: 10.3390/nu14183715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
Background and Aims: A compromise in intestinal mucosal functions is associated with several chronic inflammatory diseases. Previously, we reported that obese humans have a reduced expression of intestinal Janus kinase-3 (Jak3), a non-receptor tyrosine kinase, and a deficiency of Jak3 in mice led to predisposition to obesity-associated metabolic syndrome. Since meta-analyses show cognitive impairment as co-morbidity of obesity, the present study demonstrates the mechanistic role of Jak3 in obesity associated cognitive impairment. Our data show that high-fat diet (HFD) suppresses Jak3 expression both in intestinal mucosa and in the brain of wild-type mice. Methodology: Recapitulating these conditions using global (Jak3-KO) and intestinal epithelial cell-specific conditional (IEC-Jak3-KO) mice and using cognitive testing, western analysis, flow cytometry, immunofluorescence microscopy and 16s rRNA sequencing, we demonstrate that HFD-induced Jak3 deficiency is responsible for cognitive impairments in mice, and these are, in part, specifically due to intestinal epithelial deficiency of Jak3. Results: We reveal that Jak3 deficiency leads to gut dysbiosis, compromised TREM-2-functions-mediated activation of microglial cells, increased TLR-4 expression and HIF1-α-mediated inflammation in the brain. Together, these lead to compromised microglial-functions-mediated increased deposition of β-amyloid (Aβ) and hyperphosphorylated Tau (pTau), which are responsible for cognitive impairments. Collectively, these data illustrate how the drivers of obesity promote cognitive impairment and demonstrate the underlying mechanism where HFD-mediated impact on IEC-Jak3 deficiency is responsible for Jak3 deficiency in the brain, reduced microglial TREM2 expression, microglial activation and compromised clearance of Aβ and pTau as the mechanism during obesity-associated cognitive impairments. Conclusion: Thus, we not only demonstrate the mechanism of obesity-associated cognitive impairments but also characterize the tissue-specific role of Jak3 in such conditions through mucosal tolerance, gut–brain axis and regulation of microglial functions.
Collapse
|
13
|
Hardy-Sosa A, León-Arcia K, Llibre-Guerra JJ, Berlanga-Acosta J, Baez SDLC, Guillen-Nieto G, Valdes-Sosa PA. Diagnostic Accuracy of Blood-Based Biomarker Panels: A Systematic Review. Front Aging Neurosci 2022; 14:683689. [PMID: 35360215 PMCID: PMC8963375 DOI: 10.3389/fnagi.2022.683689] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 01/24/2022] [Indexed: 01/10/2023] Open
Abstract
Background Because of high prevalence of Alzheimer's disease (AD) in low- and middle-income countries (LMICs), there is an urgent need for inexpensive and minimally invasive diagnostic tests to detect biomarkers in the earliest and asymptomatic stages of the disease. Blood-based biomarkers are predicted to have the most impact for use as a screening tool and predict the onset of AD, especially in LMICs. Furthermore, it has been suggested that panels of markers may perform better than single protein candidates. Methods Medline/Pubmed was searched to identify current relevant studies published from January 2016 to December 2020. We included all full-text articles examining blood-based biomarkers as a set of protein markers or panels to aid in AD's early diagnosis, prognosis, and characterization. Results Seventy-six articles met the inclusion criteria for systematic review. Majority of the studies reported plasma and serum as the main source for biomarker determination in blood. Protein-based biomarker panels were reported to aid in AD diagnosis and prognosis with better accuracy than individual biomarkers. Conventional (amyloid-beta and tau) and neuroinflammatory biomarkers, such as amyloid beta-42, amyloid beta-40, total tau, phosphorylated tau-181, and other tau isoforms, were the most represented. We found the combination of amyloid beta-42/amyloid beta-40 ratio and APOEε4 status to be most represented with high accuracy for predicting amyloid beta-positron emission tomography status. Conclusion Assessment of Alzheimer's disease biomarkers in blood as a non-invasive and cost-effective alternative will potentially contribute to early diagnosis and improvement of therapeutic interventions. Given the heterogeneous nature of AD, combination of markers seems to perform better in the diagnosis and prognosis of the disease than individual biomarkers.
Collapse
Affiliation(s)
- Anette Hardy-Sosa
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
- Centro de Ingeniería Genética y Biotecnología, La Habana, Cuba
| | | | | | | | - Saiyet de la C. Baez
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
- Centro de Ingeniería Genética y Biotecnología, La Habana, Cuba
| | | | - Pedro A. Valdes-Sosa
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
- Centro de Neurociencias de Cuba, La Habana, Cuba
| |
Collapse
|
14
|
Fontana L, Ghezzi L, Cross AH, Piccio L. Effects of dietary restriction on neuroinflammation in neurodegenerative diseases. J Exp Med 2021; 218:211666. [PMID: 33416892 PMCID: PMC7802371 DOI: 10.1084/jem.20190086] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/29/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
Recent and accumulating work in experimental animal models and humans shows that diet has a much more pervasive and prominent role than previously thought in modulating neuroinflammatory and neurodegenerative mechanisms leading to some of the most common chronic central nervous system (CNS) diseases. Chronic or intermittent food restriction has profound effects in shaping brain and peripheral metabolism, immunity, and gut microbiome biology. Interactions among calorie intake, meal frequency, diet quality, and the gut microbiome modulate specific metabolic and molecular pathways that regulate cellular, tissue, and organ homeostasis as well as inflammation during normal brain aging and CNS neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis, among others. This review discusses these findings and their potential application to the prevention and treatment of CNS neuroinflammatory diseases and the promotion of healthy brain aging.
Collapse
Affiliation(s)
- Luigi Fontana
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.,Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, Australia.,Department of Clinical and Experimental Sciences, Brescia University, Brescia, Italy
| | - Laura Ghezzi
- Department of Neurology, Washington University in St. Louis, St. Louis, MO.,University of Milan, Milan, Italy
| | - Anne H Cross
- Department of Neurology, Washington University in St. Louis, St. Louis, MO
| | - Laura Piccio
- Department of Neurology, Washington University in St. Louis, St. Louis, MO.,Brain and Mind Centre, School of Medical Sciences, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
15
|
Tyagi A, Pugazhenthi S. Targeting Insulin Resistance to Treat Cognitive Dysfunction. Mol Neurobiol 2021; 58:2672-2691. [PMID: 33483903 DOI: 10.1007/s12035-021-02283-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 01/05/2021] [Indexed: 02/06/2023]
Abstract
Dementia is a devastating disease associated with aging. Alzheimer's disease is the most common form of dementia, followed by vascular dementia. In addition to clinically diagnosed dementia, cognitive dysfunction has been reported in diabetic patients. Recent studies are now beginning to recognize type 2 diabetes mellitus, characterized by chronic hyperglycemia and insulin resistance, as a risk factor for Alzheimer's disease and other cognitive disorders. While studies on insulin action have remained traditionally in the domain of peripheral tissues, the detrimental effects of insulin resistance in the central nervous system on cognitive dysfunction are increasingly being reported by recent clinical and preclinical studies. The findings from these studies suggest that antidiabetic drugs have the potential to be used to treat dementia. In this review, we discuss the physiological functions of insulin in the brain, studies on the evaluation of cognitive function under conditions of insulin resistance, and reports on the beneficial actions of antidiabetic drugs in the brain. This review covers clinical studies as well as investigations in animal models and will further highlight the emerging link between insulin resistance and neurodegenerative disorders.
Collapse
Affiliation(s)
- Anit Tyagi
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA.,Department of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA.,University of Denver, Denver, CO, USA
| | - Subbiah Pugazhenthi
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA. .,Department of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
16
|
Abstract
Brain insulin signaling contributes to memory function and might be a viable target in the prevention and treatment of memory impairments including Alzheimer's disease. This short narrative review explores the potential of central nervous system (CNS) insulin administration via the intranasal pathway to improve memory performance in health and disease, with a focus on the most recent results. Proof-of-concept studies and (pilot) clinical trials in individuals with mild cognitive impairment or Alzheimer's disease indicate that acute and prolonged intranasal insulin administration enhances memory performance, and suggest that brain insulin resistance is a pathophysiological factor in Alzheimer's disease with or without concomitant metabolic dysfunction. Intranasally administered insulin is assumed to trigger improvements in synaptic plasticity and regional glucose uptake as well as alleviations of Alzheimer's disease neuropathology; additional contributions of changes in hypothalamus-pituitary-adrenocortical axis activity and sleep-related mechanisms are discussed. While intranasal insulin delivery has been conclusively demonstrated to be effective and safe, the recent outcomes of large-scale clinical studies underline the need for further investigations, which might also yield new insights into sex differences in the response to intranasal insulin and contribute to the optimization of delivery devices to grasp the full potential of intranasal insulin for Alzheimer's disease.
Collapse
Affiliation(s)
- Manfred Hallschmid
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Otfried-Müller-Str. 25, 72076, Tübingen, Germany.
- German Center for Diabetes Research (DZD), Tübingen, Germany.
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany.
| |
Collapse
|