651
|
Secnik J, Schwertner E, Alvarsson M, Hammar N, Fastbom J, Winblad B, Garcia-Ptacek S, Religa D, Eriksdotter M. Cholinesterase inhibitors in patients with diabetes mellitus and dementia: an open-cohort study of ~23 000 patients from the Swedish Dementia Registry. BMJ Open Diabetes Res Care 2020; 8:8/1/e000833. [PMID: 31958305 PMCID: PMC7039592 DOI: 10.1136/bmjdrc-2019-000833] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/24/2019] [Accepted: 11/28/2019] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE Cholinesterase inhibitors (ChEIs) and memantine are the only approved pharmacological treatments for Alzheimer's disease (AD). Recent literature suggests reductions in cardiovascular burden and risk of stroke in ChEI users. However, the clinical effectiveness of these drugs in patients with diabetes mellitus (DM) and dementia has not been evaluated. RESEARCH DESIGN AND METHODS We conducted a registry-based open-cohort study of 22 660 patients diagnosed with AD and mixed-pathology dementia registered in the Swedish Dementia Registry until December 2015. Information on drug use, comorbidity and mortality was extracted using the linkage with the National Patient Registry, the Prescribed Drug Registry and the Cause of Death Registry. In total, 3176 (14%) patients with DM and 19 484 patients without DM were identified. Propensity-score matching, Cox-regression and competing-risk regression models were applied to produce HRs with 95% CIs for differences in all-cause, cardiovascular and diabetes-related mortality rates in ChEI users and non-users. RESULTS After matching the ChEI use in patients with DM was associated with 24% all-cause mortality reduction (HR 0.76 (95% CI 0.67 to 0.86)), compared with 20% reduction (0.80 (0.75 to 0.84)) in non-DM users. Donepezil and galantamine use were associated with a reduced mortality in both patients with DM (0.84 (0.74 to 0.96); 0.80 (0.66 to 0.97)) and patients without DM (0.85 (0.80 to 0.90); 0.93 (0.86 to 0.99)). Donepezil was further associated with reduction in cardiovascular mortality, however only in patients without DM (0.84 (0.75 to 0.94)). Rivastigmine lowered mortality only in the whole-cohort analysis and in patients without DM (0.82 (0.75 to 0.89)). Moreover, ChEI use was associated with 48% reduction in diabetes-related mortality (HR 0.52 (0.32 to 0.87)) in the whole-cohort analysis. Last, low and high doses were associated with similar benefit. CONCLUSIONS We found reductions in mortality in patients with DM and AD or mixed-pathology dementia treated with ChEIs, specifically donepezil and galantamine were associated with largest benefit. Future studies should evaluate whether ChEIs help maintain self-management of diabetes in patients with dementia.
Collapse
Affiliation(s)
- Juraj Secnik
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Huddinge, Sweden
| | - Emilia Schwertner
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Huddinge, Sweden
| | - Michael Alvarsson
- Growth and Metabolism, Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Niklas Hammar
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Johan Fastbom
- Aging Research Center, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Bengt Winblad
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
- Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Sara Garcia-Ptacek
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Huddinge, Sweden
- Theme Aging, Karolinska University Hospital, Huddinge, Sweden
- Section for Neurology, Department of Internal Medicine, Södersjukhuset - Stockholm South General Hospital, Stockholm, Sweden
| | - Dorota Religa
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Huddinge, Sweden
- Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Maria Eriksdotter
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Huddinge, Sweden
- Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
652
|
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
|
653
|
Kumari A, Sharma R, Shrivastava N, Somvanshi P, Grover A. Bleomycin modulates amyloid aggregation in β-amyloid and hIAPP. RSC Adv 2020; 10:25929-25946. [PMID: 35518630 PMCID: PMC9055351 DOI: 10.1039/d0ra04949b] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 06/29/2020] [Indexed: 12/06/2022] Open
Abstract
Aberrant misfolding and amyloid aggregation, which result in amyloid fibrils, are frequent and critical pathological incidents in various neurodegenerative disorders. Multiple drugs or inhibitors have been investigated to avert amyloid aggregation in individual peptides, exhibiting sequence-dependent inhibition mechanisms. Establishing or inventing inhibitors capable of preventing amyloid aggregation in a wide variety of amyloid peptides is quite a daunting task. Bleomycin (BLM), a complex glycopeptide, has been widely used as an antibiotic and antitumor drug due to its ability to inhibit DNA metabolism, and as an antineoplastic, especially for solid tumors. In this study, we investigated the dual inhibitory effects of BLM on Aβ aggregation, associated with Alzheimer's disease and hIAPP, which is linked to type 2 diabetes, using both computational and experimental techniques. Combined results from drug repurposing and replica exchange molecular dynamics simulations demonstrate that BLM binds to the β-sheet region considered a hotspot for amyloid fibrils of Aβ and hIAPP. BLM was also found to be involved in β-sheet destabilization and, ultimately, in its reduction. Further, experimental validation through in vitro amyloid aggregation assays was obtained wherein the fibrillar load was decreased for the BLM-treated Aβ and hIAPP peptides in comparison to controls. For the first time, this study shows that BLM is a dual inhibitor of Aβ and hIAPP amyloid aggregation. In the future, the conformational optimization and processing of BLM may help develop various efficient sequence-dependent inhibitors against amyloid aggregation in various amyloid peptides. Bleomycin acts as a dual inhibitor against both amyloid β and human islet amyloid polypeptide by binding to the β-sheet grooves considered as the amyloids hotspot.![]()
Collapse
Affiliation(s)
- Anchala Kumari
- Department of Biotechnology
- Teri School of Advanced Studies
- New Delhi
- India
- School of Biotechnology
| | - Ritika Sharma
- School of Biotechnology
- Jawaharlal Nehru University
- New Delhi
- India
| | | | - Pallavi Somvanshi
- Department of Biotechnology
- Teri School of Advanced Studies
- New Delhi
- India
| | - Abhinav Grover
- School of Biotechnology
- Jawaharlal Nehru University
- New Delhi
- India
| |
Collapse
|
654
|
Schalkwijk CG, Stehouwer CDA. Methylglyoxal, a Highly Reactive Dicarbonyl Compound, in Diabetes, Its Vascular Complications, and Other Age-Related Diseases. Physiol Rev 2020; 100:407-461. [DOI: 10.1152/physrev.00001.2019] [Citation(s) in RCA: 176] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The formation and accumulation of methylglyoxal (MGO), a highly reactive dicarbonyl compound, has been implicated in the pathogenesis of type 2 diabetes, vascular complications of diabetes, and several other age-related chronic inflammatory diseases such as cardiovascular disease, cancer, and disorders of the central nervous system. MGO is mainly formed as a byproduct of glycolysis and, under physiological circumstances, detoxified by the glyoxalase system. MGO is the major precursor of nonenzymatic glycation of proteins and DNA, subsequently leading to the formation of advanced glycation end products (AGEs). MGO and MGO-derived AGEs can impact on organs and tissues affecting their functions and structure. In this review we summarize the formation of MGO, the detoxification of MGO by the glyoxalase system, and the biochemical pathways through which MGO is linked to the development of diabetes, vascular complications of diabetes, and other age-related diseases. Although interventions to treat MGO-associated complications are not yet available in the clinical setting, several strategies to lower MGO have been developed over the years. We will summarize several new directions to target MGO stress including glyoxalase inducers and MGO scavengers. Targeting MGO burden may provide new therapeutic applications to mitigate diseases in which MGO plays a crucial role.
Collapse
Affiliation(s)
- C. G. Schalkwijk
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, The Netherlands; and Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - C. D. A. Stehouwer
- CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, The Netherlands; and Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
655
|
Bi T, Zhan L, Zhou W, Sui H. Effect of the ZiBuPiYin Recipe on Diabetes-Associated Cognitive Decline in Zucker Diabetic Fatty Rats After Chronic Psychological Stress. Front Psychiatry 2020; 11:272. [PMID: 32372981 PMCID: PMC7186306 DOI: 10.3389/fpsyt.2020.00272] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/20/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Cognitive impairment is a complication of type 2 diabetes mellitus (T2DM) that affects the central nervous system (CNS). Studies have shown that chronic psychological stress may promote the development of T2DM into diabetes-associated cognitive decline (DACD). Previously, cognitive impairment in T2DM was correlated predominantly with insulin resistance in the medial prefrontal cortex (mPFC). AIMS We examined the effect of the ZiBuPiYin recipe (ZBPYR) on Zucker diabetic fatty (ZDF) rats and explored the impact of chronic stress on altered β-amyloid (Aβ) metabolism through insulin receptor substrate (IRS) 1/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway after the induction of chronic psychological stress. MAIN METHODS After chronic psychological stress and drug treatment, cognitive function was observed via behavioral experiments. The activation of the hypothalamus-pituitary-adrenal (HPA) axis and levels of Aβ were detected by enzyme-linked immunosorbent assay, and the expression of related proteins was evaluated by Western blotting. KEY FINDINGS ZBPYR treatment significantly decreased anxious-like behaviors and plasma corticosterone (CORT) levels, and ameliorated learning and memory impairments of ZDF rats after chronic psychological stress. ZBPYR also reduced the deposition of Aβ in the mPFC, improved brain insulin resistance, and modulated the mTOR-autophagy pathway. SIGNIFICANCE ZBPYR may be a potential therapeutic application for the treatment of DACD induced by chronic psychological stress.
Collapse
Affiliation(s)
- Tingting Bi
- Modern Research Laboratory of Spleen Visceral Manifestations Theory, School of Traditional Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Libin Zhan
- Modern Research Laboratory of Spleen Visceral Manifestations Theory, School of Traditional Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wen Zhou
- Modern Research Laboratory of Spleen Visceral Manifestations Theory, School of Traditional Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hua Sui
- Institute of Integrative Medicine, Dalian Medical University, Dalian, China
| |
Collapse
|
656
|
De Sousa Rodrigues ME, Houser MC, Walker DI, Jones DP, Chang J, Barnum CJ, Tansey MG. Targeting soluble tumor necrosis factor as a potential intervention to lower risk for late-onset Alzheimer's disease associated with obesity, metabolic syndrome, and type 2 diabetes. ALZHEIMERS RESEARCH & THERAPY 2019; 12:1. [PMID: 31892368 PMCID: PMC6937979 DOI: 10.1186/s13195-019-0546-4] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 10/16/2019] [Indexed: 02/08/2023]
Abstract
Background Insulin impairment and inflammation are two features common to type 2 diabetes and Alzheimer’s disease; however, the molecular and signaling interactions underlying this relationship are not well understood. Mounting evidence point to the associations between the disruption of metabolite processing in insulin impairment and neurodegenerative conditions such as Alzheimer’s. Although the brain depends partially on metabolites processed in the periphery, to date, little is known about how soluble tumor necrosis factor signaling (solTNF) impacts integrated peripheral immune and metabolic feedback signals in states of energy overload and insulin insensitivity. Methods C57Bl/6J mice were fed a high-fat high-carbohydrate diet (HFHC) for 14 weeks. The brain-permeant biologic XPro1595® was used to block solTNF-dependent pathways. Metabolic and immune alterations were evaluated in the gut, liver, and brain. Behavioral tests were performed. Untargeted metabolomics was carried out in the plasma and liver. Results HFHC diet promotes central insulin impairment and dysregulation of immune-modulatory gene expressed in the brain. Alteration of metabolites associated with type 2 diabetes and Alzheimer’s such as butanoate, glutamate, biopterin, branched-chain amino acids, purines, and proteoglycan metabolism was observed in HFHC-fed mice. solTNF inhibition ameliorates hepatic metabolic disturbances and hepatic and intestinal lipocalin-2 levels, and decreases insulin impairment in the brain and behavioral deficits associated with HFHC diet. Conclusions Our novel findings suggest that HFHC diet impacts central insulin signaling and immune-metabolic interactions in a solTNF-dependent manner to increase the risk for neurodegenerative conditions. Our novel findings indicate that selective solTNF neutralization can ameliorate peripheral and central diet-induced insulin impairment and identify lipocalin-2 as a potential target for therapeutic intervention to target inflammation and insulin disturbances in obesogenic environments. Collectively, our findings identify solTNF as a potential target for therapeutic intervention in inflammatory states and insulin disturbances in obesogenic environments to lower risk for AD.
Collapse
Affiliation(s)
| | - Madelyn C Houser
- Department of Physiology, School of Medicine at Emory University, 615 Michael Street, Atlanta, GA, 30322-3110, USA
| | - Douglas I Walker
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University Emory, 615 Michael Street, Atlanta, GA, 30322, USA.,Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10003, USA
| | - Dean P Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, Emory University Emory, 615 Michael Street, Atlanta, GA, 30322, USA
| | - Jianjun Chang
- Department of Physiology, School of Medicine at Emory University, 615 Michael Street, Atlanta, GA, 30322-3110, USA
| | - Christopher J Barnum
- Department of Physiology, School of Medicine at Emory University, 615 Michael Street, Atlanta, GA, 30322-3110, USA
| | - Malú G Tansey
- Department of Physiology, School of Medicine at Emory University, 615 Michael Street, Atlanta, GA, 30322-3110, USA. .,Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, McKnight Brain Institute, Gainesville, FL, USA.
| |
Collapse
|
657
|
Bhattamisra SK, Yap KH, Rao V, Choudhury H. Multiple Biological Effects of an Iridoid Glucoside, Catalpol and Its Underlying Molecular Mechanisms. Biomolecules 2019; 10:E32. [PMID: 31878316 PMCID: PMC7023090 DOI: 10.3390/biom10010032] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 12/13/2022] Open
Abstract
Catalpol, an iridoid glucoside, is widely distributed in many plant families and is primarily obtained from the root of Rehmanniaglutinosa Libosch. Rehmanniaglutinosa is a plant very commonly used in Chinese and Korean traditional medicine for various disorders, including diabetes mellitus, neuronal disorders, and inflammation. Catalpol has been studied extensively for its biological properties both in vitro and in vivo. This review aims to appraise the biological effects of catalpol and their underlying mechanisms. An extensive literature search was conducted using the keyword "Catalpol" in the public domains of Google scholar, PubMed, and Scifinder. Catalpol exhibits anti-diabetic, cardiovascular protective, neuroprotective, anticancer, hepatoprotective, anti-inflammatory, and anti-oxidant effects in experimental studies. Anti-inflammatory and antioxidant properties are mostly related for its biological effect. However, some specific mechanisms are also elucidated. Elevated serotonin and BDNF level by catalpol significantly protect against depression and neurodegeneration. Catalpol demonstrated an increased mitochondrial biogenesis and activation of PI3K/Akt pathway for insulin sensitizing effect. Further, its cardiovascular protective effect was linked to PI3K/Akt, apelin/APJ and Jak-Stat pathway. Catalpol produced a significant reduction in cell proliferation and an increase in apoptosis in different cancer conditions. Overall, catalpol demonstrated multiple biological effects due to its numerous mechanisms including anti-inflammatory and antioxidant effects.
Collapse
Affiliation(s)
- Subrat Kumar Bhattamisra
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Kah Heng Yap
- School of Post graduate studies, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia; (K.H.Y.); (V.R.)
| | - Vikram Rao
- School of Post graduate studies, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia; (K.H.Y.); (V.R.)
| | - Hira Choudhury
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia;
| |
Collapse
|
658
|
Li J, Guo Y, Li Q, Miao K, Wang C, Zhang D, Tian C, Zhang S. Presence of White Matter Lesions Associated with Diabetes-Associated Cognitive Decline in Male Rat Models of Pre-Type 2 Diabetes. Med Sci Monit 2019; 25:9679-9689. [PMID: 31848329 PMCID: PMC6930701 DOI: 10.12659/msm.918557] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The aim of this study was to determine the association between white matter lesions (WML) and diabetes-associated cognitive decline (DACD) in rat models of type 2 diabetes (T2DM). MATERIAL AND METHODS Sixty Sprague-Dawley male rats were divided into 4 groups: control, control+metformin, T2DM, and T2DM+metformin groups. The T2DM groups were fed a diet high in fat and glucose to induce impaired glucose tolerance (IGT) and then were injected with streptozotocin to induce T2DM. The Morris water maze test was used to evaluate cognitive function. Brain diffusion tensor imaging scans were performed for WML. The expression of myelin basic protein (MBP), oligodendrocyte transcription factor 1 (OLIG1), and OLIG2 (markers of brain damage and repair) was determined using immunofluorescence. After IGT, the fractional anisotropy (FA) values of the right thalamus area were significantly lower in both T2DM groups compared with controls. RESULTS Eight weeks after streptozotocin injection, the FA values of the thalamus were lower in the T2DM (bilateral thalamus) group and T2DM+metformin (left thalamus) group than in controls, while the FA values in the left thalamus area were lower in the T2DM+metformin group than in the control and control+metformin groups. The maze escape latency was longer and the number of rats passing through the platform was smaller in the T2DM and T2DM+metformin groups than in the control group. MBP levels were lower and OLIG1 and OLIG2 levels were higher in both T2DM groups than in controls. CONCLUSIONS WML is associated with DACD and appears before the onset of T2DM and signs of DACD and plays a role in diabetes-associated cognitive decline. Metformin reduces WMLs but does not rescue cognitive dysfunction.
Collapse
Affiliation(s)
- Jun Li
- Department of Endocrinology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Yafei Guo
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Qingju Li
- Department of Endocrinology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Keke Miao
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Chongxian Wang
- Department of Endocrinology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Dongming Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Chenguang Tian
- Department of Endocrinology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Suhe Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| |
Collapse
|
659
|
Gao H, Jiang Q, Ji H, Ning J, Li C, Zheng H. Type 1 diabetes induces cognitive dysfunction in rats associated with alterations of the gut microbiome and metabolomes in serum and hippocampus. Biochim Biophys Acta Mol Basis Dis 2019; 1865:165541. [DOI: 10.1016/j.bbadis.2019.165541] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/23/2019] [Accepted: 08/26/2019] [Indexed: 12/14/2022]
|
660
|
Arnoriaga-Rodríguez M, Fernández-Real JM. Microbiota impacts on chronic inflammation and metabolic syndrome - related cognitive dysfunction. Rev Endocr Metab Disord 2019; 20:473-480. [PMID: 31884557 DOI: 10.1007/s11154-019-09537-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cognitive dysfunction, one of the major concerns of increased life expectancy, is prevalent in patients with metabolic disorders. Added to the inflammation in the context of aging (inflammaging), low-grade chronic inflammation (metaflammation) accompanies metabolic diseases. Peripheral and central inflammation underlie metabolic syndrome - related cognitive dysfunction. The gut microbiota is increasingly recognized to be linked to both inflammaging and metaflammation in parallel to the pathophysiology of obesity, type 2 diabetes and the metabolic syndrome. Microbiota composition, diversity and diverse metabolites have been related to different metabolic features and cognitive traits. The study of different mouse models has contributed to identify characteristic microbiota profiles and shifts in the microbial gene richness in association with cognitive function. Diet, exercise and prebiotics, probiotics or symbiotics significantly influence cognition and changes in the microbiota. Few studies have analyzed the gut microbiota composition in association with cognitive function in humans. Impaired attention, mental flexibility and executive function have been observed in association with a microbiota ecosystem in cross-sectional and longitudinal studies. Nevertheless, the evidence in humans is still scarce and not causal relationships may be inferred, so larger and long-term studies are required to gain insight into the possible role of microbiota in human cognition.
Collapse
Affiliation(s)
- María Arnoriaga-Rodríguez
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona Biomedical Research Institute [IdibGi], Carretera de França s/n, 17007, Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain
- CIBEROBN Physiopathology of Obesity and Nutrition (CIBEROBN), Madrid, Spain
- Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona Biomedical Research Institute [IdibGi], Carretera de França s/n, 17007, Girona, Spain.
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain.
- CIBEROBN Physiopathology of Obesity and Nutrition (CIBEROBN), Madrid, Spain.
- Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain.
| |
Collapse
|
661
|
Cognitive performance in relation to hydration status and water intake among older adults, NHANES 2011–2014. Eur J Nutr 2019; 59:3133-3148. [DOI: 10.1007/s00394-019-02152-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 11/20/2019] [Indexed: 12/14/2022]
|
662
|
The Novel Perspectives of Adipokines on Brain Health. Int J Mol Sci 2019; 20:ijms20225638. [PMID: 31718027 PMCID: PMC6887733 DOI: 10.3390/ijms20225638] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022] Open
Abstract
First seen as a fat-storage tissue, the adipose tissue is considered as a critical player in the endocrine system. Precisely, adipose tissue can produce an array of bioactive factors, including cytokines, lipids, and extracellular vesicles, which target various systemic organ systems to regulate metabolism, homeostasis, and immune response. The global effects of adipokines on metabolic events are well defined, but their impacts on brain function and pathology remain poorly defined. Receptors of adipokines are widely expressed in the brain. Mounting evidence has shown that leptin and adiponectin can cross the blood–brain barrier, while evidence for newly identified adipokines is limited. Significantly, adipocyte secretion is liable to nutritional and metabolic states, where defective circuitry, impaired neuroplasticity, and elevated neuroinflammation are symptomatic. Essentially, neurotrophic and anti-inflammatory properties of adipokines underlie their neuroprotective roles in neurodegenerative diseases. Besides, adipocyte-secreted lipids in the bloodstream can act endocrine on the distant organs. In this article, we have reviewed five adipokines (leptin, adiponectin, chemerin, apelin, visfatin) and two lipokines (palmitoleic acid and lysophosphatidic acid) on their roles involving in eating behavior, neurotrophic and neuroprotective factors in the brain. Understanding and regulating these adipokines can lead to novel therapeutic strategies to counteract metabolic associated eating disorders and neurodegenerative diseases, thus promote brain health.
Collapse
|
663
|
Liu W, Zhou Y, Liang R, Zhang Y. Inhibition of cyclin-dependent kinase 5 activity alleviates diabetes-related cognitive deficits. FASEB J 2019; 33:14506-14515. [PMID: 31689375 DOI: 10.1096/fj.201901292r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cognitive deficit is a prevalent and underestimated complication of diabetes, and the underlying cellular and molecular mechanisms are not well understood. Aberrant activity of cyclin-dependent kinase (Cdk)5 is implicated in a number of neurodegenerative diseases. The present study examined the role of Cdk5 in the progression of diabetes-related cognitive deficits. We showed that the Cdk5 protein expression and kinase activity were significantly increased in diabetic mice at 16 wk. In primary cultured hippocampal neurons exposed to 30 mM glucose, Cdk5 protein and kinase activity were also elevated in a time-dependent manner. Moreover, the high glucose exposure led to an aberrant Cdk5 activation due to its activator p25 that was cleaved from p35 by calpain. Both in diabetic mice and in cultured hippocampal neurons exposed to high glucose, inhibition of Cdk5 activity with roscovitine (Ros) or short hairpin RNA (shRNA) decreased the protein levels of cleaved caspase-3 and the ratio of Bax and Bcl-2. The apoptotic rate detected by TUNEL in vivo or Annexin V and propidium iodide staining for flow cytometry in vitro also had obvious reduction. In addition, high glucose exposure resulted in the increase of phosphorylated (phospho)-MAPK kinase (MKK)6, phospho-p38, and c-Jun, which were rescued by Ros or Cdk5 shRNA. It is more important that the cognitive deficits of diabetic mice were also effectively alleviated by Ros. These results indicate that aberrant Cdk5 activity triggered hippocampal neuron apoptosis by activating MKK6/p38 MAPK cascade in hyperglycemia. Inhibition of Cdk5 overactivation attenuates neuronal apoptosis and cognitive deficits and contributes to the relief of diabetic neurotoxicity in the brain.-Liu, W., Zhou, Y., Liang, R., Zhang, Y. Inhibition of cyclin-dependent kinase 5 activity alleviates diabetes-related cognitive deficits.
Collapse
Affiliation(s)
- Wei Liu
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Yi Zhou
- Department of Neurology, The Second Hospital of Hebei Medical University, Hebei Medical University, Shijiazhuang, China
| | - Rui Liang
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Yue Zhang
- Department of Clinical Diagnostics, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
664
|
Xue M, Xu W, Ou YN, Cao XP, Tan MS, Tan L, Yu JT. Diabetes mellitus and risks of cognitive impairment and dementia: A systematic review and meta-analysis of 144 prospective studies. Ageing Res Rev 2019; 55:100944. [PMID: 31430566 DOI: 10.1016/j.arr.2019.100944] [Citation(s) in RCA: 305] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 07/10/2019] [Accepted: 08/15/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Uncertainties persist about the associations of diabetes with risk of cognitive impairment and dementia. We aimed to illuminate these associations from various aspects. METHODS We identified relevant prospective studies by searching PubMed up to Jun 2019. Summary relative risks (RR) were estimated using random-effects models. Credibility of each meta-analysis was assessed. Meta-regression and subgroup analyses were conducted. RESULTS Of 28,082 identified literatures, 144 were eligible for inclusion in the systematic review, among which 122 were included in the meta-analysis. Diabetes conferred a 1.25- to 1.91-fold excess risk for cognitive disorders (cognitive impairment and dementia). Subjects with prediabetes also had higher risk for dementia. As for diabetes-related biochemical indicators, fasting plasma glucose (FPG) was non-linearly related to cognitive disorders; the elevated levels of 2 -h postload glucose (2h-PG), glycosylated hemoglobin (HbA1c), low and high levels of fasting plasma insulin (FPI) were associated with an increased risk of dementia. Encouragingly, the use of pioglitazone exhibited a 47% reduced risk of dementia in diabetic population. CONCLUSIONS Diabetes, even prediabetes and changes of diabetes-related biochemical indicators, predicted increased incidence of cognitive impairment and dementia. The protective effects of pioglitazone warrant further investigation in randomized trials.
Collapse
Affiliation(s)
- Mei Xue
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Wei Xu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xi-Peng Cao
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Meng-Shan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
665
|
Nan F, Sun G, Xie W, Ye T, Sun X, Zhou P, Dong X, Sun J, Sun X, Zhang M. Ginsenoside Rb1 mitigates oxidative stress and apoptosis induced by methylglyoxal in SH-SY5Y cells via the PI3K/Akt pathway. Mol Cell Probes 2019; 48:101469. [PMID: 31629029 DOI: 10.1016/j.mcp.2019.101469] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/26/2019] [Accepted: 10/13/2019] [Indexed: 12/24/2022]
Abstract
Diabetic encephalopathy is a severe diabetic complication characterized by cognitive dysfunction and neuropsychiatric disability. Methylglyoxal (MGO), a highly reactive metabolite of hyperglycemia, serves as a major precursor of advanced glycation end products that play key roles in diabetic complications. Ginsenoside Rb1 (abbreviated as Rb1) has received extensive attention due to its potential therapeutic effects on diabetes and neurodegeneration. Therefore, this study aimed to investigate the effects of Rb1 on MGO-induced damage in SH-SY5Y cells and the related mechanism. SH-SY5Y cells were pretreated with Rb1 for 8 h and then exposed to MGO (0.5 mM) for 24 h. Cell survival was assessed by the MTT assay. Cell apoptosis was assessed using Hoechst 33342/propidium iodide (PI) staining and an Annexin-V/PI kit. The activities of oxidative stress markers were examined using commercial kits. Reactive oxygen species (ROS) staining and JC-1 staining were used to evaluate mitochondria injury. In addition, protein levels were measured by Western blot analysis. As a result, Rb1 alleviated the injury induced by MGO by increasing the activities of superoxide dismutase, catalase and total glutathione, decreasing the level of malondialdehyde, and alleviating mitochondrial damage and ROS production. Furthermore, Rb1 could enhance the Bcl-2/Bax ratio, inhibit the expression of cleaved caspase-3 and cleaved caspase-9, and enhance the levels of phosphorylated Akt. Moreover, the protective effects of Rb1 against MGO-induced apoptosis were partly abolished by LY294002, a specific inhibitor of phosphatidylinositol 3-kinase (PI3K) phosphorylation. Our results demonstrated that Rb1 ameliorated MGO-induced oxidative stress and apoptosis in SH-SY5Y cells via activating the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Fengwei Nan
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Guibo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China
| | - Weijie Xie
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China
| | - Tianyuan Ye
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China
| | - Xiao Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China
| | - Ping Zhou
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China
| | - Xi Dong
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China
| | - Jiafu Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China
| | - Xiaobo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100193, China
| | - Mengren Zhang
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
666
|
Borshchev YY, Uspensky YP, Galagudza MM. Pathogenetic pathways of cognitive dysfunction and dementia in metabolic syndrome. Life Sci 2019; 237:116932. [PMID: 31606384 DOI: 10.1016/j.lfs.2019.116932] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 09/27/2019] [Accepted: 10/02/2019] [Indexed: 12/13/2022]
Abstract
The prevalence of dementia worldwide is growing at an alarming rate. A number of studies and meta-analyses have provided evidence for increased risk of dementia in patients with metabolic syndrome (MS) as compared to persons without MS. However, there are some reports demonstrating a lack of association between MS and increased dementia risk. In this review, taking into account the potential role of individual MS components in the pathogenesis of MS-related cognitive dysfunction, we considered the underlying mechanisms in arterial hypertension, diabetes mellitus, dyslipidemia, and obesity. The pathogenesis of dementia in MS is multifactorial, involving both vascular injury and non-ischemic neuronal death due to neurodegeneration. Neurodegenerative and ischemic lesions do not simply coexist in the brain due to independent evolution, but rather exacerbate each other, leading to more severe consequences for cognition than would either pathology alone. In addition to universal mechanisms of cognitive dysfunction shared by all MS components, other pathogenetic pathways leading to cognitive deficits and dementia, which are specific for each component, also play a role. Examples of such component-specific pathogenetic pathways include central insulin resistance and hypoglycemia in diabetes, neuroinflammation and adipokine imbalance in obesity, as well as arteriolosclerosis and lipohyalinosis in arterial hypertension. A more detailed understanding of cognitive disorders based on the recognition of underlying molecular mechanisms will aid in the development of new methods for prevention and treatment of devastating cognitive problems in MS.
Collapse
Affiliation(s)
- Yury Yu Borshchev
- Institute of Experimental Medicine, Almazov National Medical Research Center, Saint Petersburg, Russian Federation
| | - Yury P Uspensky
- Department of Faculty Therapy, Saint Petersburg State Pediatric Medical University, Saint Petersburg, Russian Federation
| | - Michael M Galagudza
- Laboratory of Digital and Display Holography, ITMO University, Russian Federation, Saint Petersburg, Russian Federation.
| |
Collapse
|
667
|
Biessels GJ, Verhagen C, Janssen J, van den Berg E, Zinman B, Rosenstock J, George JT, Passera A, Schnaidt S, Johansen OE. Effect of Linagliptin on Cognitive Performance in Patients With Type 2 Diabetes and Cardiorenal Comorbidities: The CARMELINA Randomized Trial. Diabetes Care 2019; 42:1930-1938. [PMID: 31399442 DOI: 10.2337/dc19-0783] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 07/15/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Type 2 diabetes is associated with cognitive dysfunction and an increased dementia risk, particularly in individuals with concomitant cardiovascular and/or kidney disease. Incretin therapies may modulate this risk via glycemic and nonglycemic pathways. We explored if the dipeptidyl peptidase 4 inhibitor linagliptin could prevent cognitive decline in people with type 2 diabetes with cardiorenal disease. RESEARCH DESIGN AND METHODS The CArdiovascular and Renal Microvascular outcomE study with LINAgliptin (CARMELINA)-COG substudy was an integral part of CARMELINA (NCT01897532) that randomized participants with cardiorenal disease to linagliptin 5 mg or placebo once daily (1:1), in addition to standard of care. The primary cognitive outcome was the occurrence of accelerated cognitive decline at the end of treatment, defined as a regression-based index score ≤16th percentile on the Mini-Mental State Examination (MMSE) or a composite measure of attention and executive functioning and analyzed in participants with a baseline MMSE ≥24. Effects across subgroups by baseline factors, as well as absolute cognitive changes, were also assessed. RESULTS Of the 6,979 participants in CARMELINA, CARMELINA-COG included 1,545 (mean ± SD age, 68 ± 8 years; MMSE, 28.3 ± 1.7; estimated glomerular filtration rate, 52 ± 23 mL/min/1.73 m2; and HbA1c, 7.8 ± 0.9% [61.4 ± 10.1 mmol/mol]). Over a median treatment duration of 2.5 years, accelerated cognitive decline occurred in 28.4% (linagliptin) vs. 29.3% (placebo) (odds ratio 0.96 [95% CI 0.77, 1.19]). Consistent effects were observed across subgroups by baseline characteristics. Absolute cognitive performance changes were also similar between treatment groups. CONCLUSIONS In a large international cardiovascular outcome trial in people with type 2 diabetes and cardiorenal disease, linagliptin did not modulate cognitive decline over 2.5 years.
Collapse
Affiliation(s)
- Geert Jan Biessels
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Chloë Verhagen
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jolien Janssen
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands.,Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Esther van den Berg
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands.,Department of Neurology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Bernard Zinman
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | | | - Jyothis T George
- Therapeutic Area Cardiometabolism, Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim, Germany
| | | | - Sven Schnaidt
- Biostatistics and Data Sciences, Boehringer Ingelheim, Biberach, Germany
| | | | | |
Collapse
|
668
|
Martinez-Valbuena I, Valenti-Azcarate R, Amat-Villegas I, Riverol M, Marcilla I, de Andrea CE, Sánchez-Arias JA, Del Mar Carmona-Abellan M, Marti G, Erro ME, Martínez-Vila E, Tuñon MT, Luquin MR. Amylin as a potential link between type 2 diabetes and alzheimer disease. Ann Neurol 2019; 86:539-551. [PMID: 31376172 DOI: 10.1002/ana.25570] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Alzheimer disease (AD) is the leading cause of dementia, and although its etiology remains unclear, it seems that type 2 diabetes mellitus (T2DM) and other prediabetic states of insulin resistance could contribute to the appearance of sporadic AD. As such, we have assessed whether tau and β-amyloid (Aβ) deposits might be present in pancreatic tissue of subjects with AD, and whether amylin, an amyloidogenic protein deposited in the pancreas of T2DM patients, might accumulate in the brain of AD patients. METHODS We studied pancreatic and brain tissue from 48 individuals with no neuropathological alterations and from 87 subjects diagnosed with AD. We examined Aβ and tau accumulation in the pancreas as well as that of amylin in the brain. Moreover, we performed proximity ligation assays to ascertain whether tau and/or Aβ interact with amylin in either the pancreas or brain of these subjects. RESULTS Cytoplasmic tau and Aβ protein deposits were detected in pancreatic β cells of subjects with AD as well as in subjects with a normal neuropathological examination but with a history of T2DM and in a small cohort of control subjects without T2DM. Furthermore, we found amylin deposits in the brain of these subjects, providing histological evidence that amylin can interact with Aβ and tau in both the pancreas and hippocampus. INTERPRETATION The presence of both tau and Aβ inclusions in pancreatic β cells, and of amylin deposits in the brain, provides new evidence of a potential overlap in the mechanisms underlying the pathogenesis of T2DM and AD. ANN NEUROL 2019;86:539-551.
Collapse
Affiliation(s)
- Ivan Martinez-Valbuena
- Neurology Department, Clínica Universidad de Navarra, Pamplona, Spain
- Regenerative Therapy Laboratory, Neurosciences Division, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
- Navarra Health Research Institute, Pamplona, Spain
| | - Rafael Valenti-Azcarate
- Neurology Department, Clínica Universidad de Navarra, Pamplona, Spain
- Regenerative Therapy Laboratory, Neurosciences Division, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
- Navarra Health Research Institute, Pamplona, Spain
| | - Irene Amat-Villegas
- Navarra Health Research Institute, Pamplona, Spain
- Pathology Department, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Mario Riverol
- Neurology Department, Clínica Universidad de Navarra, Pamplona, Spain
- Navarra Health Research Institute, Pamplona, Spain
| | - Irene Marcilla
- Regenerative Therapy Laboratory, Neurosciences Division, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
- Navarra Health Research Institute, Pamplona, Spain
| | | | - Juan Antonio Sánchez-Arias
- Navarra Health Research Institute, Pamplona, Spain
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Maria Del Mar Carmona-Abellan
- Neurology Department, Clínica Universidad de Navarra, Pamplona, Spain
- Regenerative Therapy Laboratory, Neurosciences Division, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
- Navarra Health Research Institute, Pamplona, Spain
| | - Gloria Marti
- Neurology Department, Clínica Universidad de Navarra, Pamplona, Spain
- Navarra Health Research Institute, Pamplona, Spain
| | - Maria-Elena Erro
- Navarra Health Research Institute, Pamplona, Spain
- Neurology Department, Navarra Hospital Complex, Pamplona, Spain
| | - Eduardo Martínez-Vila
- Neurology Department, Clínica Universidad de Navarra, Pamplona, Spain
- Navarra Health Research Institute, Pamplona, Spain
| | - Maria-Teresa Tuñon
- Navarra Health Research Institute, Pamplona, Spain
- Pathology Department, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Maria-Rosario Luquin
- Neurology Department, Clínica Universidad de Navarra, Pamplona, Spain
- Regenerative Therapy Laboratory, Neurosciences Division, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
- Navarra Health Research Institute, Pamplona, Spain
| |
Collapse
|
669
|
Verma N, Despa F. Contributing Factors to Diabetic Brain Injury and Cognitive Decline. Diabetes Metab J 2019; 43:560-567. [PMID: 31694078 PMCID: PMC6834839 DOI: 10.4093/dmj.2019.0153] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/02/2019] [Indexed: 01/11/2023] Open
Abstract
The link of diabetes with co-occurring disorders in the brain involves complex and multifactorial pathways. Genetically engineered rodents that express familial Alzheimer's disease-associated mutant forms of amyloid precursor protein and presenilin 1 (PSEN1) genes provided invaluable insights into the mechanisms and consequences of amyloid deposition in the brain. Adding diabetes factors (obesity, insulin impairment) to these animal models to predict success in translation to clinic have proven useful at some extent only. Here, we focus on contributing factors to diabetic brain injury with the aim of identifying appropriate animal models that can be used to mechanistically dissect the pathophysiology of diabetes-associated cognitive dysfunction and how diabetes medications may influence the development and progression of cognitive decline in humans with diabetes.
Collapse
Affiliation(s)
- Nirmal Verma
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Florin Despa
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
670
|
James SN, Wong A, Tillin T, Hardy R, Chaturvedi N, Richards M. The effect of mid-life insulin resistance and type 2 diabetes on older-age cognitive state: the explanatory role of early-life advantage. Diabetologia 2019; 62:1891-1900. [PMID: 31359084 PMCID: PMC6731197 DOI: 10.1007/s00125-019-4949-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 05/28/2019] [Indexed: 12/20/2022]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes, hyperglycaemia and insulin resistance are associated with cognitive impairment and dementia, but causal inference studies using Mendelian randomisation do not confirm this. We hypothesised that early-life cognition and social/educational advantage may confound the relationship. METHODS From the population-based British 1946 birth cohort, a maximum number of 1780 participants had metabolic variables (type 2 diabetes, insulin resistance [HOMA2-IR] and HbA1c) assessed at age 60-64 years, and cognitive state (Addenbrooke's Cognitive Examination III [ACE-III]) and verbal memory assessed at age 69 years. Earlier-life measures included socioeconomic position (SEP), cognition at age 8 years and educational attainment. Polygenic risk scores (PRSs) for type 2 diabetes were calculated. We first used a PRS approach with multivariable linear regression to estimate associations between PRSs and metabolic traits and later-life cognitive state. Second, using a path model approach, we estimated the interrelationships between earlier-life measures, features of mid-life type 2 diabetes and cognitive state at age 69 years. All models were adjusted for sex. RESULTS The externally weighted PRS for type 2 diabetes was associated with mid-life metabolic traits (e.g. HOMA2-IR β = 0.08 [95% CI 0.02, 0.16]), but not with ACE-III (β = 0.04 [-0.02, 0.90]) or other cognitive outcomes. While there was an association between HOMA2-IR and subsequent ACE-III (β = -0.09 [-0.15, -0.03]), path modelling showed no direct effect (β = -0.01 [-0.06, 0.03]) after accounting for the association between childhood SEP and education with HOMA2-IR. The same pattern was observed for later-life verbal memory. CONCLUSIONS/INTERPRETATION Associations between type 2 diabetes and mid-life metabolic traits with subsequent cognitive state do not appear causal, and instead they may be explained by SEP in early life, childhood cognition and educational attainment. Therefore, glucose-lowering medication may be unlikely to combat cognitive impairment in older age.
Collapse
Affiliation(s)
- Sarah-Naomi James
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, 1-19 Torrington Place, London, WC1E 7HB, UK.
| | - Andrew Wong
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, 1-19 Torrington Place, London, WC1E 7HB, UK
| | - Therese Tillin
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, 1-19 Torrington Place, London, WC1E 7HB, UK
| | - Rebecca Hardy
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, 1-19 Torrington Place, London, WC1E 7HB, UK
| | - Nishi Chaturvedi
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, 1-19 Torrington Place, London, WC1E 7HB, UK
| | - Marcus Richards
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, 1-19 Torrington Place, London, WC1E 7HB, UK
| |
Collapse
|
671
|
Zhou P, Santoro A, Peroni OD, Nelson AT, Saghatelian A, Siegel D, Kahn BB. PAHSAs enhance hepatic and systemic insulin sensitivity through direct and indirect mechanisms. J Clin Invest 2019; 129:4138-4150. [PMID: 31449056 PMCID: PMC6763232 DOI: 10.1172/jci127092] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 06/24/2019] [Indexed: 12/30/2022] Open
Abstract
Palmitic acid esters of hydroxy stearic acids (PAHSAs) are bioactive lipids with antiinflammatory and antidiabetic effects. PAHSAs reduce ambient glycemia and improve glucose tolerance and insulin sensitivity in insulin-resistant aged chow- and high-fat diet-fed (HFD-fed) mice. Here, we aimed to determine the mechanisms by which PAHSAs improve insulin sensitivity. Both acute and chronic PAHSA treatment enhanced the action of insulin to suppress endogenous glucose production (EGP) in chow- and HFD-fed mice. Moreover, chronic PAHSA treatment augmented insulin-stimulated glucose uptake in glycolytic muscle and heart in HFD-fed mice. The mechanisms by which PAHSAs enhanced hepatic insulin sensitivity included direct and indirect actions involving intertissue communication between adipose tissue and liver. PAHSAs inhibited lipolysis directly in WAT explants and enhanced the action of insulin to suppress lipolysis during the clamp in vivo. Preventing the reduction of free fatty acids during the clamp with Intralipid infusion reduced PAHSAs' effects on EGP in HFD-fed mice but not in chow-fed mice. Direct hepatic actions of PAHSAs may also be important, as PAHSAs inhibited basal and glucagon-stimulated EGP directly in isolated hepatocytes through a cAMP-dependent pathway involving Gαi protein-coupled receptors. Thus, this study advances our understanding of PAHSA biology and the physiologic mechanisms by which PAHSAs exert beneficial metabolic effects.
Collapse
Affiliation(s)
- Peng Zhou
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Anna Santoro
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Odile D. Peroni
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew T. Nelson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, California, USA
| | - Alan Saghatelian
- Clayton Foundation Laboratories for Peptide Biology, Helmsley Center for Genomic Medicine, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Dionicio Siegel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, California, USA
| | - Barbara B. Kahn
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
672
|
Takenoshita N, Shimizu S, Kanetaka H, Sakurai H, Suzuki R, Miwa T, Odawara M, Ishii K, Shimada H, Higuchi M, Suhara T, Hanyu H. Classification of Clinically Diagnosed Alzheimer’s Disease Associated with Diabetes Based on Amyloid and Tau PET Results. J Alzheimers Dis 2019; 71:261-271. [DOI: 10.3233/jad-190620] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Naoto Takenoshita
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Soichiro Shimizu
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Hidekazu Kanetaka
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Hirofumi Sakurai
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| | - Ryo Suzuki
- Department of Diabetes, Endocrinology and Metabolism, Tokyo Medical University, Tokyo, Japan
| | - Takashi Miwa
- Department of Diabetes, Endocrinology and Metabolism, Tokyo Medical University, Tokyo, Japan
| | - Masato Odawara
- Department of Diabetes, Endocrinology and Metabolism, Tokyo Medical University, Tokyo, Japan
| | - Kenji Ishii
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Hitoshi Shimada
- Department of Functional Brain Imaging Research, Clinical Research Cluster, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba-shi, Chiba, Japan
| | - Makoto Higuchi
- Department of Functional Brain Imaging Research, Clinical Research Cluster, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba-shi, Chiba, Japan
| | - Tetsuya Suhara
- Department of Functional Brain Imaging Research, Clinical Research Cluster, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba-shi, Chiba, Japan
| | - Haruo Hanyu
- Department of Geriatric Medicine, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
673
|
Ly H, Despa F. Diabetes-related Amylin Dyshomeostasis: a Contributing Factor to Cerebrovascular Pathology and Dementia. J Lipid Atheroscler 2019; 8:144-151. [PMID: 32821704 PMCID: PMC7379112 DOI: 10.12997/jla.2019.8.2.144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/21/2019] [Accepted: 06/04/2019] [Indexed: 01/11/2023] Open
Abstract
Type 2 diabetes (T2D) increases the risk for cerebrovascular disease (CVD) and dementia. The underlying molecular mechanisms remain elusive, which hampers the development of treatment or/and effective prevention strategies. Recent studies suggest that dyshomeostasis of amylin, a satiety hormone that forms pancreatic amyloid in patients with T2D, promotes accumulation of amylin in cerebral small blood vessels and interaction with Alzheimer's disease (AD) pathology. Overexpression of human amylin in rodents (rodent amylin does not form amyloid) leads to late-life onset T2D and neurologic deficits. In this Review, we discuss clinical evidence of amylin pathology in CVD and AD and identify critical characteristics of animal models that could help to better understand molecular mechanisms underlying the increased risk of CVD and AD in patients with prediabetes or T2D.
Collapse
Affiliation(s)
- Han Ly
- Departments of Pharmacology and Nutritional Sciences, and Neurology, University of Kentucky, Lexington, KY, USA
| | - Florin Despa
- Departments of Pharmacology and Nutritional Sciences, and Neurology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
674
|
The neuroprotection of liraglutide on diabetic cognitive deficits is associated with improved hippocampal synapses and inhibited neuronal apoptosis. Life Sci 2019; 231:116566. [DOI: 10.1016/j.lfs.2019.116566] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/05/2019] [Accepted: 06/12/2019] [Indexed: 12/27/2022]
|
675
|
[DIABDEM project: A pilot study of prevalence of cognitive impairment in diabetes mellitus in 2 Hispanic countries]. Rev Esp Geriatr Gerontol 2019; 54:339-345. [PMID: 31326101 DOI: 10.1016/j.regg.2019.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 03/28/2019] [Accepted: 04/01/2019] [Indexed: 01/16/2023]
Abstract
INTRODUCTION The prevalence of chronic noncommunicable diseases such as type 2 diabetes mellitus (T2DM) and dementia increase with ageing. In this context, an association between T2DM and cognitive impairment has been described in the literature. However, there are few studies in the Hispanic population. This research project presents a pilot study that will evaluate the feasibility of the DIABDEM project that will determine the prevalence of cognitive impairment in old people with diagnosis of T2DM in Spain and Chile. MATERIALS AND METHODS It is a observation-based pilot study, non-experimental, descriptive-comparative and cross-sectional. The sample will involve 72 participants (39 Spaniards and 33 Chileans), 65 year-old or older, men and women, community dwelling, and who have not been previously diagnosed with dementia, with or without a T2DM diagnosis. Participants will fill in a research protocol form collecting socio-demographic and clinical data, lifestyle details, and neuropsychological variables. EXPECTED RESULTS This study will evaluate the feasibility of the DIABDEM project that will determine the prevalence rate of cognitive impairment in old people with T2DM. On one hand, the aim of this study will establish risk and protectors factors potentially associated with the development of cognitive impairment in T2DM. On the other hand, it is expected to identify a specific neuropsychological profile in people with T2DM, proposing later a brief and useful neuropsychological battery in order to discriminate early cognitive impairment in people with T2DM. CONCLUSION Findings in this pilot study will obtain greater knowledge about the feasibility of the DIABDEM project, which will provide evidence about cognitive complications in T2DM.
Collapse
|
676
|
Iadecola C, Duering M, Hachinski V, Joutel A, Pendlebury ST, Schneider JA, Dichgans M. Vascular Cognitive Impairment and Dementia: JACC Scientific Expert Panel. J Am Coll Cardiol 2019; 73:3326-3344. [PMID: 31248555 PMCID: PMC6719789 DOI: 10.1016/j.jacc.2019.04.034] [Citation(s) in RCA: 401] [Impact Index Per Article: 80.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/09/2019] [Accepted: 04/23/2019] [Indexed: 02/07/2023]
Abstract
Cognitive impairment associated with aging has emerged as one of the major public health challenges of our time. Although Alzheimer's disease is the leading cause of clinically diagnosed dementia in Western countries, cognitive impairment of vascular etiology is the second most common cause and may be the predominant one in East Asia. Furthermore, alterations of the large and small cerebral vasculature, including those affecting the microcirculation of the subcortical white matter, are key contributors to the clinical expression of cognitive dysfunction caused by other pathologies, including Alzheimer's disease. This scientific expert panel provides a critical appraisal of the epidemiology, pathobiology, neuropathology, and neuroimaging of vascular cognitive impairment and dementia, and of current diagnostic and therapeutic approaches. Unresolved issues are also examined to shed light on new basic and clinical research avenues that may lead to mitigating one of the most devastating human conditions.
Collapse
Affiliation(s)
- Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York.
| | - Marco Duering
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-Universität LMU, Munich, Germany
| | - Vladimir Hachinski
- Department of Clinical Neurological Sciences, Western University, London, Ontario, Canada
| | - Anne Joutel
- Institute of Psychiatry and Neurosciences of Paris, INSERM U1266, Université Paris Descartes, Paris, France
| | - Sarah T Pendlebury
- Centre for Prevention of Stroke and Dementia, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital and the University of Oxford, Oxford, United Kingdom
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, Illinois
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-Universität LMU, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
677
|
Nam GE, Park YG, Han K, Kim MK, Koh ES, Kim ES, Lee MK, Kim B, Hong OK, Kwon HS. BMI, Weight Change, and Dementia Risk in Patients With New-Onset Type 2 Diabetes: A Nationwide Cohort Study. Diabetes Care 2019; 42:1217-1224. [PMID: 31177182 DOI: 10.2337/dc18-1667] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 03/31/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE This study examined the association between baseline BMI, percentage weight change, and the risk of dementia in patients newly diagnosed with type 2 diabetes. RESEARCH DESIGN AND METHODS Using the South Korean National Health Insurance Service-National Health Screening Cohort database, we identified 167,876 subjects aged ≥40 years diagnosed with new-onset type 2 diabetes between 2007 and 2012. Their weight changes were monitored for ∼2 years after diagnosis, with follow-up assessments occurring for an average of 3.5 years. The hazard ratios (HRs) and Bonferroni-adjusted 95% CIs of all-cause dementia, Alzheimer disease (AD), and vascular dementia were estimated using multivariable Cox proportional hazards regression models. RESULTS We identified 2,563 incident dementia cases during follow-up. Baseline BMI among patients with new-onset type 2 diabetes was inversely associated with the risk of all-cause dementia and AD, independent of confounding variables (P for trend <0.001). The percentage weight change during the 2 years after a diagnosis of type 2 diabetes showed significant U-shaped associations with the risk of all-cause dementia development (P < 0.001); the HRs of the disease increased significantly when weight loss or gain was >10% (1.34 [95% CI 1.11-1.63] and 1.38 [1.08-1.76], respectively). Additionally, weight loss >10% was associated with an increased risk of AD (HR 1.26 [95% CI 1.01-1.59]). CONCLUSIONS A lower baseline BMI was associated with increased risks of all-cause dementia and AD in patients with new-onset type 2 diabetes. Weight loss or weight gain after the diagnosis of diabetes was associated with an increased risk of all-cause dementia. Weight loss was associated with an increased risk of AD.
Collapse
Affiliation(s)
- Ga Eun Nam
- Department of Family Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yong Gyu Park
- Department of Biostatistics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyungdo Han
- Department of Biostatistics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mee Kyoung Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Sil Koh
- Division of Nephrology, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Sook Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Republic of Korea
| | - Min-Kyung Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Myongji Hospital, Hanyang University Medical Center, Gyeonggi-do, Republic of Korea
| | - Bongsung Kim
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea
| | - Oak-Kee Hong
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyuk-Sang Kwon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
678
|
Zhang Z, Zhang B, Wang X, Zhang X, Yang QX, Qing Z, Zhang W, Zhu D, Bi Y. Olfactory Dysfunction Mediates Adiposity in Cognitive Impairment of Type 2 Diabetes: Insights From Clinical and Functional Neuroimaging Studies. Diabetes Care 2019; 42:1274-1283. [PMID: 31221697 DOI: 10.2337/dc18-2584] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 04/17/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Large numbers of people with type 2 diabetes are obese. However, changes in cognition and related brain function in obese people with diabetes have not been characterized. Here, we investigated cognition, olfactory function, and odor-induced brain alterations in these patients and therapeutic effects of glucagon-like peptide 1 receptor agonists (GLP-1Ras) on their psychological behavior and olfactory networks. RESEARCH DESIGN AND METHODS Cognitive, olfactory, and odor-induced brain activation assessments were administered to 35 obese and 35 nonobese people with type 2 diabetes and 35 control subjects matched for age, sex, and education. Among them, 20 obese individuals with diabetes with inadequate glycemic control and metformin monotherapy received GLP-1Ra treatment for 3 months and were reassessed for metabolic, cognitive, olfactory, and neuroimaging changes. RESULTS Obese subjects with diabetes demonstrated lower general cognition and olfactory threshold scores, decreased left hippocampal activation, and disrupted seed-based functional connectivity with right insula compared with nonobese subjects with diabetes. Negative associations were found between adiposity and episodic memory and between fasting insulin and processing speed test time in diabetes. Mediation analyses showed that olfactory function and left hippocampus activation mediated these correlations. With 3-month GLP-1Ra treatment, obese subjects with diabetes exhibited improved Montreal Cognitive Assessment (MoCA) score, olfactory test total score, and enhanced odor-induced right parahippocampus activation. CONCLUSIONS Obese subjects with type 2 diabetes showed impaired cognition and dysfunctional olfaction and brain networks, the latter of which mediated adiposity in cognitive impairment of diabetes. GLP-1Ras ameliorated cognitive and olfactory abnormalities in obese subjects with diabetes, providing new perspectives for early diagnosis and therapeutic approaches for cognitive decrements in these patients.
Collapse
Affiliation(s)
- Zhou Zhang
- Department of Endocrinology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Bing Zhang
- Department of Radiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China.,Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Xin Wang
- Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Xin Zhang
- Department of Radiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Qing X Yang
- Center for NMR Research, Department of Radiology, Pennsylvania State University College of Medicine, Hershey, PA.,George M. Leader Foundation Alzheimer's Laboratory, Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey, PA
| | - Zhao Qing
- Department of Radiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Wen Zhang
- Department of Radiology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Dalong Zhu
- Department of Endocrinology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Yan Bi
- Department of Endocrinology, Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| |
Collapse
|
679
|
Moosecker S, Gomes P, Dioli C, Yu S, Sotiropoulos I, Almeida OFX. Activated PPARγ Abrogates Misprocessing of Amyloid Precursor Protein, Tau Missorting and Synaptotoxicity. Front Cell Neurosci 2019; 13:239. [PMID: 31263400 PMCID: PMC6584807 DOI: 10.3389/fncel.2019.00239] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/13/2019] [Indexed: 11/20/2022] Open
Abstract
Type 2 diabetes increases the risk for dementia, including Alzheimer’s disease (AD). Pioglitazone (Pio), a pharmacological agonist of the peroxisome proliferator-activated receptor γ (PPARγ), improves insulin sensitivity and has been suggested to have potential in the management of AD symptoms, albeit through mostly unknown mechanisms. We here investigated the potential of Pio to counter synaptic malfunction and loss, a characteristic of AD pathology and its accompanying cognitive deficits. Results from experiments on primary mouse neuronal cultures and a human neural cell line (SH-SY5Y) show that Pio treatment attenuates amyloid β (Aβ)-triggered the pathological (mis-) processing of amyloid precursor protein (APP) and inhibits Aβ-induced accumulation and hyperphosphorylation of Tau. These events are accompanied by increased glutamatergic receptor 2B subunit (GluN2B) levels that are causally linked with neuronal death. Further, Pio treatment blocks Aβ-triggered missorting of hyperphosphorylated Tau to synapses and the subsequent loss of PSD95-positive synapses. These latter effects of Pio are PPARγ-mediated since they are blocked in the presence of GW9662, a selective PPARγ inhibitor. Collectively, these data show that activated PPARγ buffer neurons against APP misprocessing, Tau hyperphosphorylation and its missorting to synapses and subsequently, synaptic loss. These first insights into the mechanisms through which PPARγ influences synaptic loss make a case for further exploration of the potential usefulness of PPARγ agonists in the prevention and treatment of synaptic pathology in AD.
Collapse
Affiliation(s)
- Susanne Moosecker
- Department Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Patrícia Gomes
- Life and Health Sciences Research Institute (ICVS), Medical School, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimarães, Portugal
| | - Chrysoula Dioli
- Life and Health Sciences Research Institute (ICVS), Medical School, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimarães, Portugal
| | - Shuang Yu
- Department Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Ioannis Sotiropoulos
- Life and Health Sciences Research Institute (ICVS), Medical School, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimarães, Portugal
| | - Osborne F X Almeida
- Department Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
680
|
Interferon-γ as a Potential Link between Diabetes Mellitus and Dementia. J Neurosci 2019; 39:4632-4635. [PMID: 31189539 DOI: 10.1523/jneurosci.3046-18.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 03/27/2019] [Accepted: 04/12/2019] [Indexed: 01/13/2023] Open
|
681
|
Lakstygal AM, de Abreu MS, Lifanov DA, Wappler-Guzzetta EA, Serikuly N, Alpsyshov ET, Wang D, Wang M, Tang Z, Yan D, Demin KA, Volgin AD, Amstislavskaya TG, Wang J, Song C, Alekseeva P, Kalueff AV. Zebrafish models of diabetes-related CNS pathogenesis. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:48-58. [PMID: 30476525 DOI: 10.1016/j.pnpbp.2018.11.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/18/2018] [Accepted: 11/22/2018] [Indexed: 12/12/2022]
Abstract
Diabetes mellitus (DM) is a common metabolic disorder that affects multiple organ systems. DM also affects brain processes, contributing to various CNS disorders, including depression, anxiety and Alzheimer's disease. Despite active research in humans, rodent models and in-vitro systems, the pathogenetic link between DM and brain disorders remains poorly understood. Novel translational models and new model organisms are therefore essential to more fully study the impact of DM on CNS. The zebrafish (Danio rerio) is a powerful novel model species to study metabolic and CNS disorders. Here, we discuss how DM alters brain functions and behavior in zebrafish, and summarize their translational relevance to studying DM-related CNS pathogenesis in humans. We recognize the growing utility of zebrafish models in translational DM research, as they continue to improve our understanding of different brain pathologies associated with DM, and may foster the discovery of drugs that prevent or treat these diseases.
Collapse
Affiliation(s)
- Anton M Lakstygal
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia
| | - Murilo S de Abreu
- Bioscience Institute, University of Passo Fundo (UPF), Passo Fundo, RS, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, USA
| | - Dmitry A Lifanov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia; School of Pharmacy, Southwest University, Chongqing, China
| | | | - Nazar Serikuly
- School of Pharmacy, Southwest University, Chongqing, China
| | | | - DongMei Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - MengYao Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - ZhiChong Tang
- School of Pharmacy, Southwest University, Chongqing, China
| | - DongNi Yan
- School of Pharmacy, Southwest University, Chongqing, China
| | - Konstantin A Demin
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Laboratory of Biological Psychiatry, Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Andrey D Volgin
- Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia
| | | | - JiaJia Wang
- Institute for Marine Drugs and Nutrition, Guangdong Ocean University, Zhanjiang, China; Marine Medicine Development Center, Shenzhen Institute, Guangdong Ocean University, Shenzhen, China
| | - Cai Song
- Institute for Marine Drugs and Nutrition, Guangdong Ocean University, Zhanjiang, China; Marine Medicine Development Center, Shenzhen Institute, Guangdong Ocean University, Shenzhen, China
| | - Polina Alekseeva
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Laboratory of Biological Psychiatry, Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia; Ural Federal University, Ekaterinburg, Russia; Russian Scientific Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia; ZENEREI Research Center, Slidell, LA, USA.
| |
Collapse
|
682
|
Ryan L, Hay M, Huentelman MJ, Duarte A, Rundek T, Levin B, Soldan A, Pettigrew C, Mehl MR, Barnes CA. Precision Aging: Applying Precision Medicine to the Field of Cognitive Aging. Front Aging Neurosci 2019; 11:128. [PMID: 31231204 PMCID: PMC6568195 DOI: 10.3389/fnagi.2019.00128] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 05/16/2019] [Indexed: 12/13/2022] Open
Abstract
The current "one size fits all" approach to our cognitive aging population is not adequate to close the gap between cognitive health span and lifespan. In this review article, we present a novel model for understanding, preventing, and treating age-related cognitive impairment (ARCI) based on concepts borrowed from precision medicine. We will discuss how multiple risk factors can be classified into risk categories because of their interrelatedness in real life, the genetic variants that increase sensitivity to, or ameliorate, risk for ARCI, and the brain drivers or common mechanisms mediating brain aging. Rather than providing a definitive model of risk for ARCI and cognitive decline, the Precision Aging model is meant as a starting point to guide future research. To that end, after briefly discussing key risk categories, genetic risks, and brain drivers, we conclude with a discussion of steps that must be taken to move the field forward.
Collapse
Affiliation(s)
- Lee Ryan
- Department of Psychology, College of Science, University of Arizona, Tucson, AZ, United States
| | - Meredith Hay
- Department of Physiology, University of Arizona, Tucson, AZ, United States
| | - Matt J. Huentelman
- Neurobehavioral Research Unit, Division of Neurological Disorders, Translational Genomics Research Institute (TGen), Phoenix, AZ, United States
| | - Audrey Duarte
- Center for Advanced Brain Imaging, School of Psychology, Georgia Institute of Technology, Atlanta, GA, United States
| | - Tatjana Rundek
- Clinical and Translational Research Division, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Bonnie Levin
- Neuropsychology Division, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Anja Soldan
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Corinne Pettigrew
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Matthias R. Mehl
- Department of Psychology, College of Science, University of Arizona, Tucson, AZ, United States
| | - Carol A. Barnes
- Department of Psychology, College of Science, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
683
|
Liu Y, Shi L, Song X, Shi C, Lou W, Zhang D, Wang AD, Luo L. Altered Brain Regional Homogeneity in First-Degree Relatives of Type 2 Diabetics: A functional MRI Study. Exp Clin Endocrinol Diabetes 2019; 128:737-744. [PMID: 31137069 DOI: 10.1055/a-0883-4955] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE This study aimed to investigate regional homogeneity in the first-: degree relatives of type 2 diabetes patients. METHODS Seventy-eight subjects, including 26 type 2 diabetes patients, 26 first-: degree relatives, and 26 healthy controls, were assessed. All participants underwent resting-state functional magnetic resonance imaging scanning. The estimated regional homogeneity value was used to evaluate differences in brain activities. RESULTS In first-: degree relatives, we observed significantly decreased regional homogeneity in the left anterior cingulate cortex, left insula, and bilateral temporal lobes, and increased regional homogeneity in the left superior frontal gyrus, right anterior cingulate cortex, and bilateral posterior cingulate cortex compared to healthy controls. In type 2 diabetes patients, we detected altered regional homogeneity in the left anterior cingulate cortex, left insula, bilateral posterior cingulate cortex, and several other brain regions compared to healthy controls. Both first-: degree relatives and type 2 diabetes patients showed decreased regional homogeneity in the left superior temporal gyrus, right middle temporal gyrus, left anterior cingulate cortex, left insula, and increased regional homogeneity in the left superior frontal gyrus and bilateral posterior cingulate cortex. CONCLUSION These findings suggest that altered regional homogeneity in the left anterior cingulate cortex, left insula, left superior frontal gyrus, bilateral posterior cingulate cortex, and bilateral temporal lobes might be a neuroimaging biomarker of type 2 diabetes -: related brain dysfunction.
Collapse
Affiliation(s)
- Yiyong Liu
- Medical Imaging Center, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Lin Shi
- Department of Imaging and Interventional Radiology, Research Centre for Medical Image Computing, The Chinese University of Hong Kong, China.,Chow Yuk Ho Technology Centre for Innovative Medicine, The Chinese University of Hong Kong, China
| | - Xiubao Song
- Department of Rehabilitation, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Changzheng Shi
- Medical Imaging Center, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Wutao Lou
- Department of Imaging and Interventional Radiology, Research Centre for Medical Image Computing, The Chinese University of Hong Kong, China
| | - Dong Zhang
- Medical Imaging Center, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Alan D Wang
- Auckland Bioengineering Institute, and Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.,Shenzhen SmartView MedTech Limited, Shenzhen, China
| | - Liangping Luo
- Medical Imaging Center, the First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
684
|
Wahl D, Solon-Biet SM, Cogger VC, Fontana L, Simpson SJ, Le Couteur DG, Ribeiro RV. Aging, lifestyle and dementia. Neurobiol Dis 2019; 130:104481. [PMID: 31136814 DOI: 10.1016/j.nbd.2019.104481] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 05/13/2019] [Accepted: 05/22/2019] [Indexed: 12/21/2022] Open
Abstract
Aging is the greatest risk factor for most diseases including cancer, cardiovascular disorders, and neurodegenerative disease. There is emerging evidence that interventions that improve metabolic health with aging may also be effective for brain health. The most robust interventions are non-pharmacological and include limiting calorie or protein intake, increasing aerobic exercise, or environmental enrichment. In humans, dietary patterns including the Mediterranean, Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER) and Okinawan diets are associated with improved age-related health and may reduce neurodegenerative disease including dementia. Rapamycin, metformin and resveratrol act on nutrient sensing pathways that improve cardiometabolic health and decrease the risk for age-associated disease. There is some evidence that they may reduce the risk for dementia in rodents. There is a growing recognition that improving metabolic function may be an effective way to optimize brain health during aging.
Collapse
Affiliation(s)
- Devin Wahl
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia; Aging and Alzheimers Institute, ANZAC Research Institute, Concord Clinical School/Sydney Medical School, Concord 2139, Australia.
| | - Samantha M Solon-Biet
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia; Aging and Alzheimers Institute, ANZAC Research Institute, Concord Clinical School/Sydney Medical School, Concord 2139, Australia
| | - Victoria C Cogger
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia; Aging and Alzheimers Institute, ANZAC Research Institute, Concord Clinical School/Sydney Medical School, Concord 2139, Australia
| | - Luigi Fontana
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia; School of Life and Environmental Sciences, University of Sydney, Sydney 2006, Australia
| | - David G Le Couteur
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia; Aging and Alzheimers Institute, ANZAC Research Institute, Concord Clinical School/Sydney Medical School, Concord 2139, Australia
| | - Rosilene V Ribeiro
- Charles Perkins Centre, University of Sydney, Sydney 2006, Australia; School of Life and Environmental Sciences, University of Sydney, Sydney 2006, Australia
| |
Collapse
|
685
|
Review of the Effect of Natural Compounds and Extracts on Neurodegeneration in Animal Models of Diabetes Mellitus. Int J Mol Sci 2019; 20:ijms20102533. [PMID: 31126031 PMCID: PMC6566911 DOI: 10.3390/ijms20102533] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/17/2019] [Accepted: 05/18/2019] [Indexed: 12/14/2022] Open
Abstract
Diabetes mellitus is a chronic metabolic disease with a high prevalence in the Western population. It is characterized by pancreas failure to produce insulin, which involves high blood glucose levels. The two main forms of diabetes are type 1 and type 2 diabetes, which correspond with >85% of the cases. Diabetes shows several associated alterations including vascular dysfunction, neuropathies as well as central complications. Brain alterations in diabetes are widely studied; however, the mechanisms implicated have not been completely elucidated. Diabetic brain shows a wide profile of micro and macrostructural changes, such as neurovascular deterioration or neuroinflammation leading to neurodegeneration and progressive cognition dysfunction. Natural compounds (single isolated compounds and/or natural extracts) have been widely assessed in metabolic disorders and many of them have also shown antioxidant, antiinflamatory and neuroprotective properties at central level. This work reviews natural compounds with brain neuroprotective activities, taking into account several therapeutic targets: Inflammation and oxidative stress, vascular damage, neuronal loss or cognitive impairment. Altogether, a wide range of natural extracts and compounds contribute to limit neurodegeneration and cognitive dysfunction under diabetic state. Therefore, they could broaden therapeutic alternatives to reduce or slow down complications associated with diabetes at central level.
Collapse
|
686
|
Darsalia V, Johansen OE, Lietzau G, Nyström T, Klein T, Patrone C. Dipeptidyl Peptidase-4 Inhibitors for the Potential Treatment of Brain Disorders; A Mini-Review With Special Focus on Linagliptin and Stroke. Front Neurol 2019; 10:493. [PMID: 31139140 PMCID: PMC6518970 DOI: 10.3389/fneur.2019.00493] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 04/23/2019] [Indexed: 12/21/2022] Open
Abstract
Cerebral stroke is a leading cause of death and persistent disability of elderly in the world. Although stroke prevention by targeting several risk factors such as diabetes and hypertension has decreased the stroke incidence, the total number of strokes is increasing due to the population aging and new preventive therapies are needed. Moreover, post-stroke acute pharmacological strategies aimed to reduce stroke-induced brain injury have failed in clinical trials despite being effective in animal models. Finally, approximately 30% of surviving stroke patients do not recover from stroke and remain permanently dependent on supportive care in activities of daily living. Therefore, strategies to improve stroke recovery in the post-acute phase are highly needed. Linagliptin is a dipeptidyl peptidase-4 inhibitor which is clinically approved to reduce hyperglycemia in type 2 diabetes. The regulation of glycemia by dipeptidyl peptidase-4 inhibition is mainly achieved by preventing endogenous glucagon-like peptide-1 (GLP-1) degradation. Interestingly, linagliptin has also shown glycaemia-independent beneficial effects in animal models of stroke, Parkinson's disease and Alzheimer's disease. In some case the preclinical data have been supported with some clinical data. Although potentially very interesting for the development of new strategies against stroke and neurodegenerative disorders, the mode of action of linagliptin in the brain is still largely unknown and seems to occur in a GLP-1R-independent manner. The purpose of this mini-review is to summarize and discuss the recent experimental and clinical work regarding the effects of linagliptin in the central nervous system, with special emphasis on acute neuroprotection, stroke prevention and post-stroke recovery. We also highlight the main questions in this research field that need to be addressed in clinical perspective.
Collapse
Affiliation(s)
- Vladimer Darsalia
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Grazyna Lietzau
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Nyström
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Klein
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Cesare Patrone
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
687
|
Intranasal insulin therapy reverses hippocampal dendritic injury and cognitive impairment in a model of HIV-associated neurocognitive disorders in EcoHIV-infected mice. AIDS 2019; 33:973-984. [PMID: 30946151 PMCID: PMC6457131 DOI: 10.1097/qad.0000000000002150] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Almost half of HIV-positive people on antiretroviral therapy have demonstrable mild neurocognitive impairment (HIV-NCI), even when virologically suppressed. Intranasal insulin therapy improves cognition in Alzheimer's disease and diabetes. Here we tested intranasal insulin therapy in a model of HIV-NCI in EcoHIV-infected conventional mice. DESIGN AND METHODS Insulin pharmacokinetics following intranasal administration to mice was determined by ELISA. Mice were inoculated with EcoHIV to cause NCI; 23 days or 3 months after infection they were treated daily for 9 days with intranasal insulin (2.4 IU/mouse) and examined for NCI in behavioral tests and HIV burdens by quantitative PCR. Some animals were tested for hippocampal neuronal integrity by immunostaining and expression of neuronal function-related genes by real time-quantitative PCR. The effect of insulin treatment discontinuation on cognition and neuropathology was also examined. RESULTS Intranasal insulin administration to mice resulted in μIU/ml levels of insulin in cerebrospinal fluid with a half-life of about 2 h, resembling pharmacokinetic parameters of patients receiving 40 IU. Intranasal insulin treatment starting 23 days or 3 months after infection completely reversed NCI in mice. Murine NCI correlated with reductions in hippocampal dendritic arbors and downregulation of neuronal function genes; intranasal insulin reversed these changes coincident with restoration of cognitive acuity, but they returned within 24 h of treatment cessation. Intranasal insulin treatment reduced brain HIV DNA when started 23 but not 90 days after infection. CONCLUSION Our preclinical studies support the use of intranasal insulin administration for treatment of HIV-NCI and suggest that some dendritic injury in this condition is reversible.
Collapse
|
688
|
|
689
|
Dichgans M. Dementia risk after transient ischaemic attack and stroke. Lancet Neurol 2019; 18:223-225. [PMID: 30784546 DOI: 10.1016/s1474-4422(18)30497-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 12/10/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Martin Dichgans
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität, Feodor-Lynen-Straße 17, D-81377, Munich, Germany; German Center for Neurodegenerative Diseases, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
690
|
Dysregulation of Glycogen Metabolism with Concomitant Spatial Memory Dysfunction in Type 2 Diabetes: Potential Beneficial Effects of Chronic Exercise. ADVANCES IN NEUROBIOLOGY 2019; 23:363-383. [DOI: 10.1007/978-3-030-27480-1_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
691
|
Huang X, Tong Y, Qi CX, Xu YT, Dan HD, Shen Y. Disrupted topological organization of human brain connectome in diabetic retinopathy patients. Neuropsychiatr Dis Treat 2019; 15:2487-2502. [PMID: 31695385 PMCID: PMC6717727 DOI: 10.2147/ndt.s214325] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 08/03/2019] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE There is increasing neuroimaging evidence that type 2 diabetes patients with retinal microvascular complications show abnormal brain functional and structural architecture and are at an increased risk of cognitive decline and dementia. However, changes in the topological properties of the functional brain connectome in diabetic retinopathy (DR) patients remain unknown. The aim of this study was to explore the topological organization of the brain connectome in DR patients using graph theory approaches. METHODS Thirty-five DR patients (18 males and 17 females) and 38 healthy controls (HCs) (18 males and 20 females), matched for age, sex, and education, underwent resting-state magnetic resonance imaging scans. Graph theory analysis was performed to investigate the topological properties of brain functional connectome at both global and nodal levels. RESULTS Both DR and HC groups showed high-efficiency small-world network in their brain functional networks. Notably, the DR group showed reduction in the clustering coefficient (P=0.0572) and local efficiency (P=0.0151). Furthermore, the DR group showed reduced nodal centralities in the default-mode network (DMN) and increased nodal centralities in the visual network (VN) (P<0.01, Bonferroni-corrected). The DR group also showed abnormal functional connections among the VN, DMN, salience network (SN), and sensorimotor network (SMN). Altered network metrics and nodal centralities were significantly correlated with visual acuity and fasting blood glucose level in DR patients. CONCLUSION DR patients showed abnormal topological organization of the human brain connectome. Specifically, the DR group showed reduction in the clustering coefficient and local efficiency, relative to HC group. Abnormal nodal centralities and functional disconnections were mainly located in the DMN, VN, SN, and SMN in DR patients. Furthermore, the disrupted topological attributes showed correlations with clinical variables. These findings offer important insight into the neural mechanism of visual loss and cognitive deficits in DR patients.
Collapse
Affiliation(s)
- Xin Huang
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, People's Republic of China
| | - Yan Tong
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, People's Republic of China
| | - Chen-Xing Qi
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, People's Republic of China
| | - Yang-Tao Xu
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, People's Republic of China
| | - Han-Dong Dan
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, People's Republic of China
| | - Yin Shen
- Eye Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, People's Republic of China
| |
Collapse
|
692
|
Bello-Chavolla OY, Antonio-Villa NE, Vargas-Vázquez A, Ávila-Funes JA, Aguilar-Salinas CA. Pathophysiological Mechanisms Linking Type 2 Diabetes and Dementia: Review of Evidence from Clinical, Translational and Epidemiological Research. Curr Diabetes Rev 2019; 15:456-470. [PMID: 30648514 DOI: 10.2174/1573399815666190129155654] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/26/2018] [Accepted: 01/08/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Type 2 diabetes represents an increasing health burden world-wide and its prevalence in particularly higher in elderly population. Consistent epidemiological evidence suggests an increased risk of dementia associated to type 2 diabetes; the mechanisms underlying these associations, however, remain unclear. OBJECTIVE The study aims to review epidemiological, clinical and pre-clinical data that weigh on pathophysiological links, mechanisms of disease and associations between type 2 diabetes and dementia to identify areas of opportunity for future research. METHODS We searched the following electronic bibliographic databases: PUBMED, EMBASE, SCIELO, MEDLINE and OVID for clinical, translational and epidemiological research literature that summarize diabetes-related risk factors for dementia, metabolic and neurological changes associated to T2D, evidence of therapeutic approaches in type 2 diabetes and its pathophysiological implications for dementia. RESULTS Type 2 diabetes mellitus increases risk for all-cause dementia, vascular dementia and Alzheimer's disease. The most evaluated mechanisms linking both disorders in pre-clinical studies include an increase in neuronal insulin resistance, impaired insulin signaling, pro-inflammatory state, mitochondrial dysfunction and vascular damage which increase deposition of β-amyloid, tau proteins and GSK3β, leading to an earlier onset of dementia in individuals with impairment in the glucose metabolism. Neuroimaging and neuropathology evidence linking cerebrovascular lesions, neurodegeneration and particularly small-vessel disease in the onset of dementia is consistent with the increased risk of incident dementia in type 2 diabetes, but consistent evidence of AD-related pathology is scarce. Epidemiological data shows increased risk of dementia related to hypoglycemic episodes, glycemic control, metabolic syndrome, insulin resistance and genetic predisposition, but the evidence is not consistent and statistical analysis might be affected by inconsistent covariate controlling. Therapeutic approaches for T2D have shown inconsistent result in relation to dementia prevention and delay of cognitive decline; lifestyle intervention, particularly physical activity, is a promising alternative to ameliorate the impact of disability and frailty on T2D-related dementia. CONCLUSION Vascular disease, inflammation and impaired brain insulin signaling might occur in T2D and contribute to dementia risk. Evidence from epidemiological studies has not consistently reported associations that could integrate a unified mechanism of disease in humans. Evaluation of the effect of antidiabetic medications and non-pharmacological interventions in dementia prevention in type 2 diabetes is promising but has thus far offered inconsistent results.
Collapse
Affiliation(s)
- Omar Yaxmehen Bello-Chavolla
- Metabolic Disease Research Unit, National Institute of Medical Sciences and Nutrition, Mexico, United States
- Faculty of Medicine, National Autonomous, University of Mexico, Mexico, United States
| | - Neftali Eduardo Antonio-Villa
- Metabolic Disease Research Unit, National Institute of Medical Sciences and Nutrition, Mexico, United States
- Faculty of Medicine, National Autonomous, University of Mexico, Mexico, United States
| | - Arsenio Vargas-Vázquez
- Metabolic Disease Research Unit, National Institute of Medical Sciences and Nutrition, Mexico, United States
- Faculty of Medicine, National Autonomous, University of Mexico, Mexico, United States
| | - José Alberto Ávila-Funes
- Department of Geriatrics, National Institute of Medical Sciences and Nutrition, Mexico, United States
- University of Bordeaux, Bordeaux Population Health Research Center, 33076 Bordeaux, France
| | - Carlos Alberto Aguilar-Salinas
- Department of Endocrinology and Metabolism, National Institute of Medical Sciences and Nutrition, Mexico, United States
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Sertoma, 64710 Monterrey, N.L., Mexico, United States
| |
Collapse
|
693
|
Ye T, Meng X, Wang R, Zhang C, He S, Sun G, Sun X. Gastrodin Alleviates Cognitive Dysfunction and Depressive-Like Behaviors by Inhibiting ER Stress and NLRP3 Inflammasome Activation in db/db Mice. Int J Mol Sci 2018; 19:ijms19123977. [PMID: 30544722 PMCID: PMC6321309 DOI: 10.3390/ijms19123977] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 12/06/2018] [Accepted: 12/07/2018] [Indexed: 12/15/2022] Open
Abstract
Patients with diabetes mellitus (DM) suffer more risks from diabetic encephalopathy such as cognitive dysfunction and depressive-like behaviors. Numerous studies show that ER (endoplasmic reticulum) stress and inflammation play important roles in the development of diabetic encephalopathy. Gastrodin (Gas), one major component of Gastrodia elata, is traditionally used in central nervous system disorders and is believed to possess anti-inflammatory, anti-apoptotic, and other neuroprotective effects. This present study aims to explore the protective effects of Gas on diabetic encephalopathy. Gas was administrated daily (70 and 140 mg/Kg) for 12 weeks. Meanwhile, the fasting blood glucose and body weight of db/db mice were measured every two weeks. After Gas treatment, the Morris water maze (MWM) test and novel object recognition (NOR) test were performed to assess the learning and memory functions of db/db mice, and the forced swim test was performed to evaluate depressive-like behaviors of db/db mice. Additionally, the expression of ER stress and Nucleotide binding and oligomerization domain-like (Nod) receptor family pyrin domain-containing 3 (NLRP3) inflammasome related proteins were evaluated by using Western blot. Our study suggested that Gas attenuated blood glucose levels and dyslipidemia of db/db mice. It has been shown that Gas could improve learning and memory function and depressive-like behaviors of db/db mice. Moreover, Gas inhibited ER stress and NLRP3 inflammasome activation in the hippocampus. Taken together, this study demonstrates that Gas attenuates the diabetic encephalopathy by inhibiting ER stress and NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Tianyuan Ye
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China.
| | - Xiangbao Meng
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China.
| | - Ruiying Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China.
| | - Chenyang Zhang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China.
| | - Shuaibing He
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China.
| | - Guibo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China.
| | - Xiaobo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing, China.
| |
Collapse
|
694
|
Groeneveld O, Reijmer Y, Heinen R, Kuijf H, Koekkoek P, Janssen J, Rutten G, Kappelle L, Biessels G. Brain imaging correlates of mild cognitive impairment and early dementia in patients with type 2 diabetes mellitus. Nutr Metab Cardiovasc Dis 2018; 28:1253-1260. [PMID: 30355471 DOI: 10.1016/j.numecd.2018.07.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/07/2018] [Accepted: 07/24/2018] [Indexed: 11/21/2022]
Abstract
BACKGROUND AND AIMS The risk of mild cognitive impairment and dementia is increased in type 2 diabetes mellitus (T2DM). We aimed to identify the neuroanatomical correlates of mild cognitive impairment (MCI) and early dementia in patients with T2DM, using advanced multimodal MRI. METHODS AND RESULTS Twenty-five patients (≥70 years) with T2DM and MCI (n = 22) or early dementia (n = 3) were included. The reference group consisted of 23 patients with T2DM with intact cognition. All patients underwent a 3 T MRI. Brain volumes and white matter hyperintensity volumes were obtained with automated segmentation methods. White matter connectivity was assessed with diffusion tensor imaging and fiber tractography. Infarcts and microbleeds were rated visually. Compared to patients without cognitive impairment, those with impairment had a lower grey matter volume (effect size: -0.58, p=0.042), especially in the right temporal lobe and subcortical brain regions (effect sizes: -0.45 to -0.91, false discovery rate corrected p < 0.05). White matter volume (effect size: -0.47, p = 0.11) and white matter connectivity (effect size: 0.55, p = 0.054) were also reduced in patients with versus without cognitive impairment, albeit not statistically significant. White matter hyperintensity volumes and occurrence of other vascular lesions did not differ between the two patient groups. CONCLUSION In patients with T2DM, grey matter atrophy rather than vascular brain injury appears to be the primary imaging correlate of MCI and early dementia.
Collapse
Affiliation(s)
- O Groeneveld
- Brain Center Rudolf Magnus, Department of Neurology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands.
| | - Y Reijmer
- Brain Center Rudolf Magnus, Department of Neurology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - R Heinen
- Brain Center Rudolf Magnus, Department of Neurology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - H Kuijf
- Image Sciences Institute, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - P Koekkoek
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - J Janssen
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - G Rutten
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - L Kappelle
- Brain Center Rudolf Magnus, Department of Neurology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - G Biessels
- Brain Center Rudolf Magnus, Department of Neurology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| |
Collapse
|
695
|
Ferreira LSS, Fernandes CS, Vieira MNN, De Felice FG. Insulin Resistance in Alzheimer's Disease. Front Neurosci 2018; 12:830. [PMID: 30542257 PMCID: PMC6277874 DOI: 10.3389/fnins.2018.00830] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/23/2018] [Indexed: 12/22/2022] Open
Abstract
The epidemiological connection between diabetes, obesity, and dementia represents an important public health challenge but also an opportunity to further understand these conditions. The key intersection among the three diseases is insulin resistance, which has been classically described to occur in peripheral tissues in diabetes and obesity and has recently been shown to develop in Alzheimer's disease (AD) brains. Here we review encouraging preclinical and clinical data indicating the potential of targeting impaired insulin signaling with antidiabetic drugs to treat dementia. We further discuss biological mechanisms through which peripheral metabolic dysregulation may lead to brain malfunction, providing possible explanations for the connection between diabetes, obesity, and AD. Finally, we briefly discuss how lifelong allostatic load may interact with aging to increase the risk of dementia in late life.
Collapse
Affiliation(s)
- Laís S. S. Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Caroline S. Fernandes
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo N. N. Vieira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda G. De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Biomedical and Molecular Sciences, Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| |
Collapse
|