1
|
Ahmed AI, AbuHaweeleh MN, Abdelhamid A, Al-Dali Y, Al-Suwaidi H, Khaled Y, Chivese T, Djouhri L. Hyperglycemia is associated with poorer cognitive performance in a cohort of middle-aged people in Qatar: a cross-sectional study. Expert Rev Endocrinol Metab 2025; 20:211-219. [PMID: 40103391 DOI: 10.1080/17446651.2025.2473407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 02/11/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Diabetes mellitus (DM) prevalence in Qatar is among the highest worldwide. DM has been shown to be associated with reduced performance on numerous domains of cognitive function in elderly population. Here, we sought to determine whether such association also exists in a middle-aged cohort. RESEARCH DESIGN AND METHODS A cross-sectional study was conducted using data from 981 participants aged 40-65 years from the Qatar Biobank. We analyzed glycemic indices: HbA1c, serum glucose, insulin levels, waist circumference, and waist-hip ratio. Cognitive function was assessed using two domains of CANTAB: the paired episodic memory (visual memory) and reaction time (motor and mental speed). RESULTS We found significant associations between DM and cognitive impairment. Poor reaction speed was linked to DM (beta 36.80, P < 0.01), higher HbA1c levels (beta 10.73, P < 0.05), larger waist circumference (beta 1.70, P < 0.001), and higher waist-to-hip ratio (beta 252.56, P ≤ 0.01). Poor memory performance was also associated with increased waist circumference and waist-to-hip ratio. CONCLUSION The negative association between DM, its biomarkers, and cognitive impairment reported previously in elderly populations also exists in middle-aged individuals. Further research is needed to explore the causality and impact of dysglycemia on other cognitive domains.
Collapse
Affiliation(s)
- Ashraf I Ahmed
- Department of Basic Medical Science, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | - Aya Abdelhamid
- Department of Basic Medical Science, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Yazan Al-Dali
- Department of Basic Medical Science, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Hissa Al-Suwaidi
- Department of Basic Medical Science, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Yousef Khaled
- Department of Basic Medical Science, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Tawanda Chivese
- Division of Science and Mathematics, School of Interdisciplinary Arts and Sciences, University of Washington, Tacoma, USA
| | - Laiche Djouhri
- Department of Basic Medical Science, College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
2
|
Sumbul‐Sekerci B, Velioglu HA, Sekerci A. Diabetes-related clinical and microstructural white matter changes in patients with Alzheimer's disease. Brain Behav 2024; 14:e3533. [PMID: 38715429 PMCID: PMC11077244 DOI: 10.1002/brb3.3533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 05/12/2024] Open
Abstract
AIM Although there exists substantial epidemiological evidence indicating an elevated risk of dementia in individuals with diabetes, our understanding of the neuropathological underpinnings of the association between Type-2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) remains unclear. This study aims to unveil the microstructural brain changes associated with T2DM in AD and identify the clinical variables contributing to these changes. METHODS In this retrospective study involving 64 patients with AD, 31 individuals had concurrent T2DM. The study involved a comparative analysis of diffusion tensor imaging (DTI) images and clinical features between patients with and without T2DM. The FSL FMRIB software library was used for comprehensive preprocessing and tractography analysis of DTI data. After eddy current correction, the "bedpost" model was utilized to model diffusion parameters. Linear regression analysis with a stepwise method was used to predict the clinical variables that could lead to microstructural white matter changes. RESULTS We observed a significant impairment in the left superior longitudinal fasciculus (SLF) among patients with AD who also had T2DM. This impairment in patients with AD and T2DM was associated with an elevation in creatine levels. CONCLUSION The white matter microstructure in the left SLF appears to be sensitive to the impairment of kidney function associated with T2DM in patients with AD. The emergence of AD in association with T2DM may be driven by mechanisms distinct from the typical AD pathology. Compromised renal function in AD could potentially contribute to impaired white matter integrity.
Collapse
Affiliation(s)
- Betul Sumbul‐Sekerci
- Department of Clinical Pharmacy, Faculty of PharmacyBezmialem Vakıf UniversityIstanbulTurkey
| | - Halil Aziz Velioglu
- Center for Psychiatric NeuroscienceFeinstein Institutes for Medical ResearchManhassetNew YorkUSA
- Department of Neuroscience, Faculty of MedicineIstanbul Medipol UniversityIstanbulTurkey
| | - Abdusselam Sekerci
- Department of Internal Medicine, Faculty of MedicineBezmialem Vakif UniversityIstanbulTurkey
| |
Collapse
|
3
|
Canário NS, Crisóstomo J, Moreno C, Duarte JV, Duarte IC, Ribeiro MJ, Caramelo B, Gomes LV, Matafome P, Oliveira FP, Castelo-Branco M. Functional reorganization of memory processing in the hippocampus is associated with neuroprotector GLP-1 levels in type 2 diabetes. Heliyon 2024; 10:e27412. [PMID: 38509913 PMCID: PMC10950584 DOI: 10.1016/j.heliyon.2024.e27412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 02/22/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
Type 2 diabetes (T2D) often impairs memory functions, suggesting specific vulnerability of the hippocampus. In vivo neuroimaging studies relating encoding and retrieval of memory information with endogenous neuroprotection are lacking. The neuroprotector glucagon-like peptide (GLP-1) has a high receptor density in anterior/ventral hippocampus, as shown by animal models. Using an innovative event-related fMRI design in 34 participants we investigated patterns of hippocampal activity in T2D (n = 17) without mild cognitive impairment (MCI) versus healthy controls (n = 17) during an episodic memory task. We directly measured neurovascular coupling by estimating the hemodynamic response function using event-related analysis related to encoding and retrieval of episodic information in the hippocampus. We applied a mixed-effects general linear model analysis and a two-factor ANOVA to test for group differences. Significant between-group differences were found for memory encoding, showing evidence for functional reorganization: T2D patients showed an augmented activation in the posterior hippocampus while anterior activation was reduced. The latter was negatively correlated with both GLP-1 pre- and post-breakfast levels, in the absence of grey matter changes. These results suggest that patients with T2D without MCI have pre-symptomatic functional reorganization in brain regions underlying episodic memory, as a function of the concentration of the neuroprotective neuropeptide GLP-1.
Collapse
Affiliation(s)
- Nádia S. Canário
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute of Nuclear Sciences Applied to Health, Portugal
- Faculty of Medicine, University of Coimbra, Portugal
| | - Joana Crisóstomo
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute of Nuclear Sciences Applied to Health, Portugal
| | - Carolina Moreno
- Department of Endocrinology, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
| | - João V. Duarte
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute of Nuclear Sciences Applied to Health, Portugal
- Faculty of Medicine, University of Coimbra, Portugal
| | - Isabel C. Duarte
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute of Nuclear Sciences Applied to Health, Portugal
| | - Mário J. Ribeiro
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute of Nuclear Sciences Applied to Health, Portugal
- The Faculty of Science and Technology, University of Coimbra, Portugal
| | - Beatriz Caramelo
- Faculty of Medicine, University of Coimbra, Portugal
- Coimbra Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine and Center of Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal
| | - Leonor V. Gomes
- Department of Endocrinology, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal
| | - Paulo Matafome
- Faculty of Medicine, University of Coimbra, Portugal
- Coimbra Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine and Center of Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal
| | | | - Miguel Castelo-Branco
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute of Nuclear Sciences Applied to Health, Portugal
- Faculty of Medicine, University of Coimbra, Portugal
| |
Collapse
|
4
|
Patil RS, Tupe RS. Communal interaction of glycation and gut microbes in diabetes mellitus, Alzheimer's disease, and Parkinson's disease pathogenesis. Med Res Rev 2024; 44:365-405. [PMID: 37589449 DOI: 10.1002/med.21987] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 07/12/2023] [Accepted: 08/06/2023] [Indexed: 08/18/2023]
Abstract
Diabetes and its complications, Alzheimer's disease (AD), and Parkinson's disease (PD) are increasing gradually, reflecting a global threat vis-à-vis expressing the essentiality of a substantial paradigm shift in research and remedial actions. Protein glycation is influenced by several factors, like time, temperature, pH, metal ions, and the half-life of the protein. Surprisingly, most proteins associated with metabolic and neurodegenerative disorders are generally long-lived and hence susceptible to glycation. Remarkably, proteins linked with diabetes, AD, and PD share this characteristic. This modulates protein's structure, aggregation tendency, and toxicity, highlighting renovated attention. Gut microbes and microbial metabolites marked their importance in human health and diseases. Though many scientific shreds of evidence are proposed for possible change and dysbiosis in gut flora in these diseases, very little is known about the mechanisms. Screening and unfolding their functionality in metabolic and neurodegenerative disorders is essential in hunting the gut treasure. Therefore, it is imperative to evaluate the role of glycation as a common link in diabetes and neurodegenerative diseases, which helps to clarify if modulation of nonenzymatic glycation may act as a beneficial therapeutic strategy and gut microbes/metabolites may answer some of the crucial questions. This review briefly emphasizes the common functional attributes of glycation and gut microbes, the possible linkages, and discusses current treatment options and therapeutic challenges.
Collapse
Affiliation(s)
- Rahul Shivaji Patil
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Rashmi Santosh Tupe
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Pune, Maharashtra, India
| |
Collapse
|
5
|
Yamashima T, Seike T, Oikawa S, Kobayashi H, Kido H, Yanagi M, Yamamiya D, Li S, Boontem P, Mizukoshi E. Hsp70.1 carbonylation induces lysosomal cell death for lifestyle-related diseases. Front Mol Biosci 2023; 9:1063632. [PMID: 36819480 PMCID: PMC9936620 DOI: 10.3389/fmolb.2022.1063632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/28/2022] [Indexed: 02/05/2023] Open
Abstract
Alzheimer's disease, type 2 diabetes, and non-alcoholic steatohepatitis (NASH) constitute increasingly prevalent disorders. Individuals with type 2 diabetes are well-known to be susceptible to Alzheimer's disease. Although the pathogenesis of each disorder is multifactorial and the causal relation remains poorly understood, reactive oxygen species (ROS)-induced lipid and protein oxidation conceivably plays a common role. Lipid peroxidation product was recently reported to be a key factor also for non-alcoholic steatohepatitis, because of inducing hepatocyte degeneration/death. Here, we focus on implication of the representative lipid-peroxidation product 'hydroxynonenal' for the cell degeneration/death of brain, pancreas, and liver. Since Hsp70.1 has dual roles as a chaperone and lysosomal membrane stabilizer, hydroxynonenal-mediated oxidative injury (carbonylation) of Hsp70.1 was highlighted. After intake of high-fat diets, oxidation of free fatty acids in mitochondria generates ROS which enhance oxidation of ω-6 polyunsaturated fatty acids (PUFA) involved within biomembranes and generate hydroxynonenal. In addition, hydroxynonenal is generated during cooking deep-fried foods with vegetable oils especially containing linoleic acids. These intrinsic and exogenous hydroxynonenal synergically causes an increase in its serum and organ levels to induce Hsp70.1 oxidation. As it is amphiphilic; being water-soluble but displays strong lipophilic characteristics, hydroxynonenal can diffuse within the cells and react with targets like senile and/or atheromatous plaques outside the cells. Hydroxynonenal can deepen and expand lysosomal injuries by facilitating 'calpain-mediated cleavage of the carbonylated Hsp70.1'. Despite the unique anatomical, physiological, and biochemical characteristics of each organ for its specific disease, there should be a common cascade of the cell degeneration/death which is caused by hydroxynonenal. This review aims to implicate hydroxynonenal-mediated Hsp70.1 carbonylation for lysosomal membrane permeabilization/rupture and the resultant cathepsin leakage for inducing cell degeneration/death. Given the tremendous number of worldwide people suffering various lifestyle-related diseases, it is valuable to consider how ω-6 PUFA-rich vegetable oils is implicated for the organ disorder.
Collapse
Affiliation(s)
- Tetsumori Yamashima
- Department of Psychiatry and Behavioral Science, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan,Department of Cell Metabolism and Nutrition, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan,*Correspondence: Tetsumori Yamashima,
| | - Takuya Seike
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Shinji Oikawa
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hatasu Kobayashi
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hidenori Kido
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Masahiro Yanagi
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Daisuke Yamamiya
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Shihui Li
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Piyakarn Boontem
- Department of Cell Metabolism and Nutrition, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Eishiro Mizukoshi
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| |
Collapse
|
6
|
Du XL, Song L. A Large Retrospective Cohort Study on the Risk of Alzheimer's Disease and Related Dementias in Association with Vascular Diseases and Cancer Therapy in Men with Prostate Cancer. J Prev Alzheimers Dis 2023; 10:193-206. [PMID: 36946446 PMCID: PMC10709824 DOI: 10.14283/jpad.2023.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND No study was conducted on the long-term risk of Alzheimer's disease (AD) and related dementias (ADRD) in association with vascular diseases in men with prostate cancer. OBJECTIVES To determine the 26-year risk of ADRD in association with cardiovascular disease (CVD), stroke, hypertension, and diabetes in a nationwide cohort of men with prostate cancer. DESIGN Retrospective cohort study. SETTING Surveillance, Epidemiology, and End Results (SEER) areas of the United States. PARTICIPANTS 351,571 men diagnosed with prostate cancer at age ≥65 years. MEASUREMENTS Main exposures were CVD, stroke, hypertension, and diabetes. Main outcome was the incidence of ADRD. RESULTS The crude 26-year cumulative incidence of any ADRD was higher in those with versus without CVD (33.80% vs 29.11%), stroke (40.70% vs 28.03%), hypertension (30.88% vs 27.31%), and diabetes (32.23% vs 28.68%). Men with CVD (adjusted hazard ratio: 1.17, 95% CI: 1.15-1.20), stroke (1.59, 1.56-1.61), hypertension (1.13, 1.11-1.14), and diabetes (1.25, 1.23-1.27) were significantly more likely to develop ADRD than those without. Patients with 4 of these vascular diseases were 161% more likely to develop ADRD (2.61, 2.47-2.76) than those without. The risk of AD (0.89, 0.87-0.91) and ADRD (0.91, 0.90-0.93) became significantly lower in men with prostate cancer who received androgen deprivation therapy as compared to those who did not after considering death as a competing risk. CONCLUSIONS In men with prostate cancer, vascular diseases were associated with significantly higher risks of developing ADRD. Androgen deprivation therapy was associated with a significantly decreased risk of AD in men with prostate cancer.
Collapse
Affiliation(s)
- X L Du
- Xianglin L. Du, PhD, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, 1200 Pressler St, Houston, TX 77030, USA;
| | | |
Collapse
|
7
|
Liu C, Guo X. Adjuvant Chinese Medicine for the Treatment of Type 2 Diabetes Mellitus Combined with Mild Cognitive Impairment: A Systematic Review and Meta-Analysis of a Randomised Controlled Trial. Pharmaceuticals (Basel) 2022; 15:ph15111424. [PMID: 36422553 PMCID: PMC9697494 DOI: 10.3390/ph15111424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/10/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022] Open
Abstract
Mild cognitive impairment has a high prevalence in the type 2 diabetic population. Adjuvant therapy with Chinese herbal medicine can effectively improve the clinical symptoms of patients with T2DM combined with MCI. The aim of this study was to systematically evaluate the efficacy and safety of Chinese herbal adjunctive therapy in the treatment of diabetes mellitus combined with cognitive impairment. Information was analysed using the China Knowledge Network, Vip Database, Wanfang Database, China Biomedical Literature Database, PubMed, EMbase, Web of Science, and MedLine Database. The total clinical efficiency, blood glucose, blood lipids, Simple Mental-State Examination Scale (MMSE), Montreal Cognitive Assessment Scale (MoCA), Traditional Chinese Medicine Symptom Score (TCMSS), and incidence of adverse reactions were recorded. The methodological quality of the included studies was evaluated using the application of the Cochrane Collaboration Network Risk Bias Assessment Tool, and meta-analysis was performed using RevMan 5.4 software. Adjuvant treatment with Chinese herbal medicine was effective in improving the clinical outcomes (OR = 5.33, 95% CI (3.62, 7.84), p < 0.00001) and cognitive function by comparing with the control group: MMSE (MD = 1.56, 95% CI (1.29, 1.84), p < 0.00001) and MoCA (MD = 2.77, 95% CI (1.81, 3.73), p < 0.0001); lowered blood glucose: fasting blood glucose (FBG) (MD = −0.27, 95% CI (−0.42, −0.12), p = 0.0006), 2 hPG (MD = −0.28, 95% CI (−0.45, −0.10), p = 0.002), and glycated haemoglobin (HbA1c) (MD = −0.26, 95% CI (−0.39, −0.14), p < 0.001); and improved lipids: total cholesterol (TC) (MD = −0.51, 95% CI (−0.82, −0.21), p = 0.001), triglycerides (TGs) (MD = −0.46, 95% CI −0.46, 95% CI (−0.80, −0.11), p = 0.009), low-density lipoprotein (LDL-C) (MD = −0.28, 95% CI (−0.55, −0.02), p = 0.04), high-density lipoprotein (HDL-C) (MD = 0.17, 95% CI (0.07, 0.28), p = 0.001), reduced TCMSS (MD = −1.84, 95% CI (−2.58, −1.10), p < 0.0001), and incidence of adverse events (OR = 0.46, 95% CI (0.24, 0.88), p = 0.02). In conclusion, through the available evidence, herbal adjuvant therapy for T2DM combined with MCI was observed to be effective and did not significantly increase the adverse effects. Due to the limitation of the number and quality of the included studies, the abovementioned results need to be validated by further high-quality studies.
Collapse
|
8
|
Relationship between the Responsiveness of Amyloid β Protein to Platelet Activation by TRAP Stimulation and Brain Atrophy in Patients with Diabetes Mellitus. Int J Mol Sci 2022; 23:ijms232214100. [PMID: 36430576 PMCID: PMC9697742 DOI: 10.3390/ijms232214100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/11/2022] [Accepted: 11/11/2022] [Indexed: 11/17/2022] Open
Abstract
Type 2 DM is a risk factor for dementia, including Alzheimer's disease (AD), and is associated with brain atrophy. Amyloid β protein (Aβ) deposition in the brain parenchyma is implicated in the neurodegeneration that occurs in AD. Platelets, known as abundant storage of Aβ, are recognized to play important roles in the onset and progression of AD. We recently showed that Aβ negatively regulates platelet activation induced by thrombin receptor-activating protein (TRAP) in healthy people. In the present study, we investigated the effects of Aβ on the TRAP-stimulated platelet activation in DM patients, and the relationship between the individual responsiveness to Aβ and quantitative findings of MRI, the volume of white matter hyperintensity (WMH)/intracranial volume (IC) and the volume of parenchyma (PAR)/IC. In some DM patients, Aβ reduced platelet aggregation induced by TRAP, while in others it was unchanged or rather enhanced. The TRAP-induced levels of phosphorylated-Akt and phosphorylated-HSP27, the levels of PDGF-AB and the released phosphorylated-HSP27 correlated with the degree of platelet aggregability. The individual levels of not WMH/IC but PAR/IC was correlated with those of TRAP-stimulated PDGF-AB release. Collectively, our results suggest that the reactivity of TRAP-stimulated platelet activation to Aβ differs in DM patients from healthy people. The anti-suppressive feature of platelet activation to Aβ might be protective for brain atrophy in DM patients.
Collapse
|
9
|
Brain structural alterations detected by an automatic quantified tool as an indicator for MCI diagnosing in type 2 diabetes mellitus patients: a magnetic resonance imaging study. Heliyon 2022; 8:e09390. [PMID: 35647347 PMCID: PMC9136264 DOI: 10.1016/j.heliyon.2022.e09390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 12/02/2021] [Accepted: 05/03/2022] [Indexed: 11/23/2022] Open
Abstract
Background and objectives Type 2 diabetes mellitus (T2DM) is an important risk factors for mild cognitive impairment (MCI). Structural magnetic resonance imaging (sMRI) is an effective and widely used method to investigate brain pathomorphological injury in neural diseases. In present study, we aimed to determine the brain regional alterations that correlated to the incidence of MCI in T2DM patients. Materials and methods Eighteen T2DM patients with and without MCI (DMCI/T2DM) respectively, and eighteen age/gender-matched healthy controls (HC) were recruited. Brain MRI imagines of all the individuals were subjected to automatic quantified brain sub-structure volume segmentation and measurement by Dr. brain ™ software. The relative volume of total gray matter (TGM), total white matter (TWM), and 68 pairs (left and right) of brain sub-structures were compared between the three groups. Cognitive function correlation analysis and receiver operating characteristic (ROC) curve analysis were conducted in the MCI-related brain regions in T2DM patients, and we utilized a machine learning method to classify the three group of subjects. Results 10 and 27 brain sub-structures with significant relative volumetric alterations were observed in T2DM patients without MCI and T2DM patients with MCI, respectively (p < 0.05). Compared with T2DM patients without MCI, eight critical regions include right anterior orbital gyrus, right calcarine and cerebrum, left cuneus, left entorhinal area, left frontal operculum, right medial orbital gyrus, right occipital pole, left temporal pole had significant lower volumetric ratio in T2DM patients with MCI (p < 0.05). Among them, the decrease of volumetric ratio in several regions had a positive correlation with Montreal Cognitive Assessment (MoCA) scores and Mini-Mental State Examination (MMSE) scores. The classification results conducted based on these regions as features by random forest algorithm yielded good accuracies of T2DM/HC 69.4%, DMCI/HC 72.2% and T2DM/DMCI 69.4%. Conclusions Certain brain regional structural lesions occurred in patients with T2DM, and this condition was more serious in T2DM patients combined with MCI. A systematic way of segmenting and measuring the whole brain has a potential clinical value for predicting the presence of MCI for T2DM patients.
Collapse
|
10
|
Lei Y, Zhang D, Qi F, Gao J, Tang M, Ai K, Yan X, Lei X, Shao Z, Su Y, Zhang X. Dysfunctional Interaction Between the Dorsal Attention Network and the Default Mode Network in Patients With Type 2 Diabetes Mellitus. Front Hum Neurosci 2022; 15:796386. [PMID: 35002661 PMCID: PMC8741406 DOI: 10.3389/fnhum.2021.796386] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/08/2021] [Indexed: 11/13/2022] Open
Abstract
The risk of cognitive impairment in patients with type 2 diabetes mellitus (T2DM) is significantly higher than that in the general population, but the exact neurophysiological mechanism underlying this is still unclear. An abnormal change in the intrinsic anticorrelation of the dorsal attention network (DAN) and the default mode network (DMN) is thought to be the mechanism underlying cognitive deficits that occur in many psychiatric disorders, but this association has rarely been tested in T2DM. This study explored the relationship between the interaction patterns of the DAN-DMN and clinical/cognitive variables in patients with T2DM. Forty-four patients with T2DM and 47 sex-, age-, and education-matched healthy controls (HCs) underwent neuropsychological assessments, independent component analysis (ICA), and functional network connection analysis (FNC). The relationship of DAN-DMN anticorrelation with the results of a battery of neuropsychological tests was also assessed. Relative to the HC group, the DMN showed decreased functional connectivity (FC) in the right precuneus, and the DAN showed decreased FC in the left inferior parietal lobule (IPL) in patients with T2DM. Subsequent FNC analysis revealed that, compared with the HC group, the T2DM patients displayed significantly increased inter-network connectivity between the DAN and DMN. These abnormal changes were correlated with the scores of multiple neuropsychological assessments (P < 0.05). These findings indicate abnormal changes in the interaction patterns of the DAN-DMN may be involved in the neuropathology of attention and general cognitive dysfunction in T2DM patients.
Collapse
Affiliation(s)
- Yumeng Lei
- Department of MRI, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Dongsheng Zhang
- Department of MRI, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Fei Qi
- Department of MRI, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Jie Gao
- Department of MRI, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Min Tang
- Department of MRI, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Kai Ai
- Department of Clinical Science, Philips Healthcare, Xi'an, China
| | - Xuejiao Yan
- Department of MRI, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Xiaoyan Lei
- Department of MRI, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Zhirong Shao
- Department of Graduate, Xi'an Medical University, Xi'an, China
| | - Yu Su
- Department of MRI, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Xiaoling Zhang
- Department of MRI, Shaanxi Provincial People's Hospital, Xi'an, China
| |
Collapse
|
11
|
O'Bryant SE, Petersen M, Hall J, Johnson L. Metabolic Factors Are Related to Brain Amyloid Among Mexican Americans: A HABS-HD Study. J Alzheimers Dis 2022; 86:1745-1750. [PMID: 35253763 PMCID: PMC9364418 DOI: 10.3233/jad-215620] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Despite the tremendous amount of research on Alzheimer's disease (AD) biomarkers, very little data is available regarding the fundamental biomarkers of AD among Mexican Americans. OBJECTIVE Here we sought to examine the link between metabolic markers and brain amyloid among Mexican Americans as compared to non-Hispanic whites from the Health & Aging Brain Study -Health Disparities (HABS-HD) cohort. METHODS PET amyloid (florbetaben) data was analyzed from 34 Mexican American and 22 non-Hispanic white participants. RESULTS Glucagon (t = 3.84, p < 0.001) and insulin (t = -2.56, p = 0.02) were both significantly related to global SUVR levels among Mexican Americans. Glucagon and insulin were both related to most ROIs. No metabolic markers were significantly related to brain amyloid levels among non-Hispanic whites. CONCLUSION Metabolic markers are related to brain amyloid burden among Mexican Americans. Given the increased risk for diabetes, additional research is needed to determine the impact of diabetes on core AD biomarkers among this underserved population.
Collapse
Affiliation(s)
- Sid E O'Bryant
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Melissa Petersen
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - James Hall
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Leigh Johnson
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
12
|
Du XL, Song L, Schulz PE, Xu H, Chan W. Associations Between Vascular Diseases and Alzheimer's Disease or Related Dementias in a Large Cohort of Men and Women with Colorectal Cancer. J Alzheimers Dis 2022; 90:211-231. [PMID: 36093703 PMCID: PMC9661325 DOI: 10.3233/jad-220548] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Long term risk of Alzheimer's disease (AD) and related dementias (ADRD) associated with vascular diseases in people with colorectal cancer is unknown. OBJECTIVE To determine the risk of ADRD in association with cardiovascular diseases (CVD), stroke, hypertension, and diabetes in a cohort of patients with colorectal cancer. METHODS This retrospective cohort study consisted of 210,809 patients diagnosed with colorectal cancer at age≥65 years in 1991-2015 from the Surveillance, Epidemiology, and End Results (SEER)-Medicare linked database with follow-up from 1991-2016, who were free of any ADRD at the baseline (<12 months prior to or < 30 days after the date of cancer diagnosis). RESULTS The crude 26-year cumulative incidence of total ADRD in men and women with colorectal cancer was higher in those with versus without CVD (31.92% versus 28.12%), with versus without stroke (39.82% versus 26.39%), with versus without hypertension (31.88% versus 24.88%), and with versus without diabetes (32.01% versus 27.66%). After adjusting for socio-demographic and tumor factors, the risk of developing ADRD was significantly higher in patients with CVD (adjusted hazard ratio: 1.17, 95% confidence intervals: 1.14-1.20), stroke (1.65, 1.62-1.68), hypertension (1.07, 1.05-1.09), and diabetes (1.26, 1.24-1.29) versus persons without. For those with 1, 2, 3 and 4 vascular diseases present versus absent, the risk of AD increased from 1.12 (1.07-1.16) to 1.31 (1.25-1.36), 1.66 (1.57-1.75), and 2.03 (1.82-2.27). CONCLUSION In older patients with colorectal cancer, a significant dose-response relationship was observed between an increasing number of these vascular diseases and the risk of all types of dementia.
Collapse
Affiliation(s)
- Xianglin L. Du
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Lulu Song
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Biostatistics and Data Science, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Paul E. Schulz
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hua Xu
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Wenyaw Chan
- Department of Biostatistics and Data Science, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
13
|
Sekar S, Viswas RS, Miranzadeh Mahabadi H, Alizadeh E, Fonge H, Taghibiglou C. Concussion/Mild Traumatic Brain Injury (TBI) Induces Brain Insulin Resistance: A Positron Emission Tomography (PET) Scanning Study. Int J Mol Sci 2021; 22:9005. [PMID: 34445708 PMCID: PMC8396497 DOI: 10.3390/ijms22169005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/04/2021] [Accepted: 08/13/2021] [Indexed: 01/07/2023] Open
Abstract
Brain injury/concussion is a growing epidemic throughout the world. Although evidence supports association between traumatic brain injury (TBI) and disturbance in brain glucose metabolism, the underlying molecular mechanisms are not well established. Previously, we reported the release of cellular prion protein (PrPc) from the brain to circulation following TBI. The PrPc level was also found to be decreased in insulin-resistant rat brains. In the present study, we investigated the molecular link between PrPc and brain insulin resistance in a single and repeated mild TBI-induced mouse model. Mild TBI was induced in mice by dropping a weight (~95 g at 1 m high) on the right side of the head. The procedure was performed once and thrice (once daily) for single (SI) and repeated induction (RI), respectively. Micro PET/CT imaging revealed that RI mice showed significant reduction in cortical, hippocampal and cerebellum glucose uptake compared to SI and control. Mice that received RI also showed significant motor and cognitive deficits. In co-immunoprecipitation, the interaction between PrPc, flotillin and Cbl-associated protein (CAP) observed in the control mice brains was disrupted by RI. Lipid raft isolation showed decreased levels of PrPc, flotillin and CAP in the RI mice brains. Based on observation, it is clear that PrPc has an interaction with CAP and the dislodgment of PrPc from cell membranes may lead to brain insulin resistance in a mild TBI mouse model. The present study generated a new insight into the pathogenesis of brain injury, which may result in the development of novel therapy.
Collapse
Affiliation(s)
- Sathiya Sekar
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada; (S.S.); (H.M.M.)
| | - Raja Solomon Viswas
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada; (R.S.V.); (E.A.)
| | - Hajar Miranzadeh Mahabadi
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada; (S.S.); (H.M.M.)
| | - Elahe Alizadeh
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada; (R.S.V.); (E.A.)
| | - Humphrey Fonge
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada; (R.S.V.); (E.A.)
- Department of Medical Imaging, Royal University Hospital (RUH), Saskatoon, SK S7N 0W8, Canada
| | - Changiz Taghibiglou
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada; (S.S.); (H.M.M.)
| |
Collapse
|
14
|
Dave N, Vural AS, Piras IS, Winslow W, Surendra L, Winstone JK, Beach TG, Huentelman MJ, Velazquez R. Identification of retinoblastoma binding protein 7 (Rbbp7) as a mediator against tau acetylation and subsequent neuronal loss in Alzheimer's disease and related tauopathies. Acta Neuropathol 2021; 142:279-294. [PMID: 33978814 PMCID: PMC8270842 DOI: 10.1007/s00401-021-02323-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/14/2021] [Accepted: 05/01/2021] [Indexed: 12/14/2022]
Abstract
Evidence indicates that tau hyper-phosphorylation and subsequent neurofibrillary tangle formation contribute to the extensive neuronal death in Alzheimer's disease (AD) and related tauopathies. Recent work has identified that increased tau acetylation can promote tau phosphorylation. Tau acetylation occurs at lysine 280 resulting from increased expression of the lysine acetyltransferase p300. The exact upstream mechanisms mediating p300 expression remain elusive. Additional work highlights the role of the epigenome in tau pathogenesis, suggesting that dysregulation of epigenetic proteins may contribute to acetylation and hyper-phosphorylation of tau. Here, we identify and focus on the histone-binding subunit of the Nucleosome Remodeling and Deacetylase (NuRD) complex: Retinoblastoma-Binding Protein 7 (Rbbp7). Rbbp7 chaperones chromatin remodeling proteins to their nuclear histone substrates, including histone acetylases and deacetylases. Notably, Rbbp7 binds to p300, suggesting that it may play a role in modulating tau acetylation. We interrogated Rbbp7 in post-mortem brain tissue, cell lines and mouse models of AD. We found reduced Rbbp7 mRNA expression in AD cases, a significant negative correlation with CERAD (neuritic plaque density) and Braak Staging (pathogenic tau inclusions) and a significant positive correlation with post-mortem brain weight. We also found a neuron-specific downregulation of Rbbp7 mRNA in AD patients. Rbbp7 protein levels were significantly decreased in 3xTg-AD and PS19 mice compared to NonTg, but no decreases were found in APP/PS1 mice that lack tau pathology. In vitro, Rbbp7 overexpression rescued TauP301L-induced cytotoxicity in immortalized hippocampal cells and primary cortical neurons. In vivo, hippocampal Rbbp7 overexpression rescued neuronal death in the CA1 of PS19 mice. Mechanistically, we found that increased Rbbp7 reduced p300 levels, tau acetylation at lysine 280 and tau phosphorylation at AT8 and AT100 sites. Collectively, these data identify a novel role of Rbbp7, protecting against tau-related pathologies, and highlight its potential as a therapeutic target in AD and related tauopathies.
Collapse
Affiliation(s)
- Nikhil Dave
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Austin S Vural
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Ignazio S Piras
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Wendy Winslow
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Likith Surendra
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Joanna K Winstone
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Thomas G Beach
- Arizona Alzheimer's Consortium, Phoenix, AZ, USA
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Matthew J Huentelman
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Ramon Velazquez
- Arizona State University-Banner Neurodegenerative Disease Research Center at the Biodesign Institute, Arizona State University, Tempe, AZ, USA.
- Arizona Alzheimer's Consortium, Phoenix, AZ, USA.
- School of Life Sciences, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
15
|
Nakasato A, Kobayashi T, Kubota M, Yamashita F, Nakaya T, Sasaki M, Kihara H, Kondo H. Increase in masseter muscle activity by newly fabricated complete dentures improved brain function. J Prosthodont Res 2021; 65:482-488. [PMID: 33762505 DOI: 10.2186/jpr.jpr_d_20_00038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
PURPOSE To reveal effects of improvement of masseter muscle activity on brain function in elderly people wearing complete dentures. METHODS Subjects were 14 edentulous patients with a chief complaint of the inconvenience of their complete dentures. The surface electromyographic (EMG) activity of the masseter muscles was measured. Brain activities were analyzed with functional magnetic resonance imaging (fMRI), employing chewing gum as the task program. Cognitive functions were evaluated with Trail Making Test Part A (TMT-A), Rey Auditory Verbal Learning Test (RAVLT) and Rey-Osterrieth Complex Figure Test (R-OCFT). Those evaluations were performed in which subjects wore their old dentures (OD) or newly fabricated dentures (ND). RESULTS We compared ND condition with OD condition. The masseter muscle activity significantly increased in ND condition (p < 0.05, Wilcoxon signed rank test). The brain activity increased significantly in the superior frontal gyrus, precentral gyrus, putamen, inferior parietal lobule, cerebellum, inferior frontal lobe, and middle frontal gyrus under the ND condition than under the OD condition (p < 0.01, uncorrected, cluster size > 10 voxels). Results of TMT-A, RAVLT, and R-OCFT were also significantly improved (p < 0.05, Wilcoxon signed rank test). CONCLUSIONS In the edentulous elderly, the brain activity was increased following the improvement of the masseter muscle activity. Consequently, it is possible that the improvement of the masseter muscle activity might influ ence on the attention, verbal skills, and visual memory.
Collapse
Affiliation(s)
- Ayaka Nakasato
- Department of Prosthodontics and Oral Implantology, School of Dentistry, Iwate Medical University, Iwate
| | - Takuya Kobayashi
- Department of Prosthodontics and Oral Implantology, School of Dentistry, Iwate Medical University, Iwate
| | - Masafumi Kubota
- Department of Prosthodontics and Oral Implantology, School of Dentistry, Iwate Medical University, Iwate
| | - Fumio Yamashita
- Division of Ultra-High Field MRI, Iwate Medical University, Iwate
| | - Takaharu Nakaya
- Faculty of Social Welfare, Iwate Prefectural University, Iwate
| | - Makoto Sasaki
- Division of Ultra-High Field MRI, Iwate Medical University, Iwate
| | - Hidemichi Kihara
- Department of Prosthodontics and Oral Implantology, School of Dentistry, Iwate Medical University, Iwate
| | - Hisatomo Kondo
- Department of Prosthodontics and Oral Implantology, School of Dentistry, Iwate Medical University, Iwate
| |
Collapse
|
16
|
Da Silva JD, Ni SC, Lee C, Elani H, Ho K, Thomas C, Kuwajima Y, Ishida Y, Kobayashi T, Ishikawa-Nagai S. Association between cognitive health and masticatory conditions: a descriptive study of the national database of the universal healthcare system in Japan. Aging (Albany NY) 2021; 13:7943-7952. [PMID: 33739304 PMCID: PMC8034966 DOI: 10.18632/aging.202843] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/05/2021] [Indexed: 12/02/2022]
Abstract
Cognitive health is subject to decline with increasing numbers of lost teeth which impacts mastication. This study is a descriptive data analysis of the association between masticatory and cognitive conditions using a large database. We obtained the dental and medical records from Japan's universal healthcare system (UHCS) from the national database in 2017. The data from 94% of the Japanese population aged 65 and over is included. It is inclusive of diagnostic codes for various types of cognitive impairment, as well as dental treatment records from 2012 to 2017. The cognitive impairment group was compared to those without a diagnosis of cognitive impairment. Crude odds ratio between loss of mastication with natural teeth (exposure) and cognitive impairments (outcome) were compared. Patients who have lost masticatory function are likely to have cognitive impairment with an odds ratio of 1.89 (p<0.0001) for early elderly (aged 65-75) and 1.33 (p<0.0001) for advanced elderly (over 75). Patients who are edentulous and function with complete dentures are likely to have cognitive impairment with an odds ratio of 2.38 (p<0.0001) and 1.38 (p<0.0001), respectively. The data shows a convincing and significant result of an association between cognitive health and oral health, related to masticatory conditions.
Collapse
Affiliation(s)
- John D Da Silva
- Restorative Dentistry and Biomaterial Sciences, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Shy Chwen Ni
- Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Cliff Lee
- Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Hawazin Elani
- Oral Health Policy and Epidemiology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Kailing Ho
- DMD Candidate, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Carlos Thomas
- DMD Candidate, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Yukinori Kuwajima
- Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Yoshiki Ishida
- Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | | | - Shigemi Ishikawa-Nagai
- Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA.,Iwate Medical University, School of Dental Medicine, Japan
| |
Collapse
|
17
|
O’Bryant SE, Zhang F, Petersen M, Johnson L, Hall J, Rissman RA. A Precision Medicine Approach to Treating Alzheimer's Disease Using Rosiglitazone Therapy: A Biomarker Analysis of the REFLECT Trials. J Alzheimers Dis 2021; 81:557-568. [PMID: 33814447 PMCID: PMC8203239 DOI: 10.3233/jad-201610] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND The REFLECT trials were conducted to examine the treatment of mild-to-moderate Alzheimer's disease utilizing a peroxisome proliferator-activated receptor gamma agonist. OBJECTIVE To generate a predictive biomarker indicative of positive treatment response using samples from the previously conducted REFLECT trials. METHODS Data were analyzed on 360 participants spanning multiple negative REFLECT trials, which included treatment with rosiglitazone and rosiglitazone XR. Support vector machine analyses were conducted to generate a predictive biomarker profile. RESULTS A pre-defined 6-protein predictive biomarker (IL6, IL10, CRP, TNFα, FABP-3, and PPY) correctly classified treatment response with 100%accuracy across study arms for REFLECT Phase II trial (AVA100193) and multiple Phase III trials (AVA105640, AV102672, and AVA102670). When the data was combined across all rosiglitazone trial arms, a global RSG-predictive biomarker with the same 6-protein predictive biomarker was able to accurately classify 98%of treatment responders. CONCLUSION A predictive biomarker comprising of metabolic and inflammatory markers was highly accurate in identifying those patients most likely to experience positive treatment response across the REFLECT trials. This study provides additional proof-of-concept that a predictive biomarker can be utilized to help with screening and predicting treatment response, which holds tremendous benefit for clinical trials.
Collapse
Affiliation(s)
- Sid E. O’Bryant
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Fan Zhang
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Melissa Petersen
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Leigh Johnson
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - James Hall
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Robert A. Rissman
- Department of Family Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
18
|
Hölscher C. Evidence for pathophysiological commonalities between metabolic and neurodegenerative diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 155:65-89. [PMID: 32854859 DOI: 10.1016/bs.irn.2020.01.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Diabetes mellitus is a risk factor for developing neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases. This relationship seems counter-intuitive as these pathological syndromes appear to be very different. However, they share underlying mechanisms such as desensitization of insulin signaling. Insulin not only regulates blood glucose levels, but also acts as a growth factor that is important for neuronal activity and repair. Insulin signaling desensitization has been found in the brains of people with progressive neurodegenerative diseases, which is most likely driven by chronic inflammation. Based on this, insulin has been tested in patients with Alzheimer's disease, and it was found that memory formation was improved and brain pathology reduced. Glucagon-like peptide-1 (GLP-1) is an incretin hormone, and numerous drugs that mimic this peptide are on the market to treat type 2 diabetes mellitus. Preclinical studies have provided robust evidence that some of these drugs, such as liraglutide or lixisenatide can enter the brain and improve key pathological parameters, such as memory loss, impairment of motor activity, synapse loss, reduced energy utilization by neurons and chronic inflammation in the brain. First clinical trials with a GLP-1 mimetic show good effects in patients with Parkinson's disease, improving motor control and insulin signaling in the brain. This is a proof of concept that this approach is viable and that drug treatment affects the main drivers of the disease and does not just modify the symptoms. It demonstrates that this new research area is a promising and fertile space for the development of novel treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Christian Hölscher
- Neurology Department of the Second Associated Hospital of Shanxi Medical University, Taiyuan, Shanxi, PR China; Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou, Henan, PR China.
| |
Collapse
|
19
|
Hölscher C. Brain insulin resistance: role in neurodegenerative disease and potential for targeting. Expert Opin Investig Drugs 2020; 29:333-348. [PMID: 32175781 DOI: 10.1080/13543784.2020.1738383] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction: This review evaluates the novel strategy of treating Alzheimer's and Parkinson's disease (AD and PD) withdrugs that initially have been developed to treat type 2 diabetes. As insulin signalling has been found to be de-sensitized in the brains of patients, drugs that can re-sensitize insulin signalling have been tested to evaluate if this strategy can alter disease progression.Areas covered: The review will give an overview of preclinical and clinical tests in AD and PD of drugs activating insulin receptors, glucagon-like peptide -1 (GLP-1) receptors, and glucose-dependent insulinotropic polypeptide (GIP) receptors.Expert opinion: Insulin, GLP-1 and GIP receptor agonists have shown good effects in preclinical studies. First clinical trials in MCI/AD patients have shown that insulin can improve on key pathological symptoms of AD such as memory impairment, brain activity, neuronal energy utilization, and inflammation markers. A GLP-1 receptor agonist has shown disease-modifying effects in PD patients, and first pilot studies have shown encouraging effects of a GLP-1 receptor agonist in AD patients. Novel dual GLP-1/GIP receptor agonists that cross the blood brain barrier show superior neuroprotective effects compared to single GLP-1 or GIP receptor agonists, and show great promise as novel treatments of AD and PD.
Collapse
Affiliation(s)
- Christian Hölscher
- Second Hospital, Neurology Department, Shanxi Medical University, Taiyuan, Shanxi, PR China.,Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou, Henan, PR China
| |
Collapse
|
20
|
Schiel JE, Spiegelhalder K. [Interaction of insomnia in old age and associated diseases : Cognitive, behavioral and neurobiological aspects]. Z Gerontol Geriatr 2020; 53:112-118. [PMID: 32020285 DOI: 10.1007/s00391-020-01694-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/19/2019] [Indexed: 12/16/2022]
Abstract
The prevalence of insomnia is particularly high in old age. Sleep disturbances and impaired daytime functioning reflected in mood swings and concentration difficulties are often accompanied by other mental disorders such as depression. The objective of this article is to shed light on the role of insomnia in the context of frequent comorbidities in middle and old age. The focus is on the identification of linkage points between insomnia and associated diseases on a neurobiological level; however, possible distinguishing features are also named and deliberations on cognitive behavioral aspects and integrative theories, such as the hyperarousal theory are discussed. In order to provide an outlook for future research opportunities, the UK Biobank is presented as a promising resource of long-term data. Finally, the contents of the preceding deliberations are critically reflected and practical implications for the treatment of older patients with insomnia are derived.
Collapse
Affiliation(s)
- J E Schiel
- Klinik für Psychiatrie und Psychotherapie, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität, Hauptstr. 6, 79104, Freiburg, Deutschland.
| | - K Spiegelhalder
- Klinik für Psychiatrie und Psychotherapie, Universitätsklinikum Freiburg, Medizinische Fakultät, Albert-Ludwigs-Universität, Hauptstr. 6, 79104, Freiburg, Deutschland
| |
Collapse
|
21
|
Wen XH, Guo QL, Guo JC. Effect of 9 - PAHSA on cognitive dysfunction in diabetic mice and its possible mechanism. Biochem Biophys Res Commun 2020; 524:525-532. [PMID: 32014256 DOI: 10.1016/j.bbrc.2020.01.071] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 01/13/2020] [Indexed: 01/19/2023]
Abstract
Diabetes mellitus (DM) is currently a major global health problem, which is associated with the development of cognitive dysfunction. However, although numerous clinical drugs for hyperglycemia have been used at present, safer and more effective therapeutic intervention strategies for diabetic cognitive impairments are still a huge challenge. Recently, several studies have indicated that a novel class of branched palmitic acid esters of hydroxyl stearic acids (PAHSAs) may have anti-diabetes and anti-inflammatory effects in insulin-resistant mice. Herein, whether the 9-PAHSA that one of the PAHSAs can attenuates DM-associated cognitive impairment in a mouse model of type 2 diabetes has been investigated. Our results showed that 9-PAHSA mildly prevented deficits of spatial working memory in Y-maze test while reversed the preference bias toward novel mice in Social choice test. Furthermore, the effect of REST on cognitive impairment of diabetes was explored for the first time. It was found that the expression of REST in diabetic mice increased, and the expression of target protein BDNF (Brain-derived neurotrophic factor) was decreased. After administration of 9-PAHSA, the situation was reversed. In summary, we conclude that exogenous supplement of 9-PAHSA can improve DM-related cognitive impairment to some extent, and the protective effect may be associated with decreased REST/NRSF (repressor element-1 silencing transcription factor/neuron-restrictive silence factor) and upregulated BDNF expression in frontal cortex.
Collapse
Affiliation(s)
- Xiao-Hong Wen
- State Key Laboratory of Medical Neurobiology & Institutes of Brain Science, Fudan University, Shanghai, China.
| | - Qi-Lin Guo
- State Key Laboratory of Medical Neurobiology & Institutes of Brain Science, Fudan University, Shanghai, China
| | - Jing-Chun Guo
- State Key Laboratory of Medical Neurobiology & Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
22
|
Sekar S, Zhang Y, Miranzadeh Mahabadi H, Parvizi A, Taghibiglou C. Low-Field Magnetic Stimulation Restores Cognitive and Motor Functions in the Mouse Model of Repeated Traumatic Brain Injury: Role of Cellular Prion Protein. J Neurotrauma 2019; 36:3103-3114. [PMID: 31020907 DOI: 10.1089/neu.2018.5918] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Traumatic brain injury (TBI)/concussion is a growing epidemic throughout the world. Memory and neurobehavioral dysfunctions are among the sequelae of TBI. Dislodgement of cellular prion protein (PrPc) and disruption of circadian rhythm have been linked to TBI. Low-field magnetic stimulation (LFMS) is a new noninvasive repetitive transcranial magnetic stimulation (rTMS) technique that generates diffused and low-intensity magnetic stimulation to deep cortical and subcortical areas. The role of LFMS on PrPc, proteins related to the circadian rhythm, and behavior alterations in a repeated TBI mouse model were studied in the present study. TBI was induced to the mice (right hemisphere) using weight-drop method, once daily for 3 days. LFMS treatment was given for 20 min once daily for 4 days (immediately after each TBI induction). The results showed that LFMS-treated TBI mice significantly improved cognitive and motor function as evidenced by open field exploration, rotarod, and novel location recognition tasks. In addition, a significant increase in PrPc and decreased glial fibrillary acidic protein levels were observed in cortical and hippocampal regions of LFMS-treated TBI mice brain compared with sham-treated TBI mice, while neuronal nuclei level was significantly increased in cortical region. In LFMS-treated mice, a decrease in proteins related to circadian rhythm were observed, compared with sham-treated TBI mice. The results obtained from the study demonstrated the neuroprotective effect of LFMS, which may be through regulating PrPc and/or proteins related to circadian rhythm. Thus, the present study suggests that LFMS may improve the subject's neurological condition following TBI.
Collapse
Affiliation(s)
- Sathiya Sekar
- Department of Anatomy, Physiology, Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Yanbo Zhang
- Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Hajar Miranzadeh Mahabadi
- Department of Anatomy, Physiology, Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Amirhassan Parvizi
- Department of Anatomy, Physiology, Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Changiz Taghibiglou
- Department of Anatomy, Physiology, Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
23
|
Rosenberg J, Lechea N, Pentang GN, Shah NJ. What magnetic resonance imaging reveals - A systematic review of the relationship between type II diabetes and associated brain distortions of structure and cognitive functioning. Front Neuroendocrinol 2019; 52:79-112. [PMID: 30392901 DOI: 10.1016/j.yfrne.2018.10.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 10/11/2018] [Accepted: 10/22/2018] [Indexed: 12/19/2022]
Abstract
Due to its increasing prevalence, Type 2 diabetes mellitus (T2DM) represents a major health challenge for modern society. Despite it being of fundamental interest, only a few MRI studies have conducted statistical analyses to draw scientifically valid conclusions about the complex interplay of T2DM and its associated clinical, structural, functional, metabolite, as well as cognitive distortions. Therefore, a systematic review of 68 manuscripts, following the PRISMA guidelines, was conducted. Notably, although the associations between imaging, clinical, and cognitive variables are not fully homogeneous, findings show a clear trend towards a link between altered brain structure and a decline in cognitive processing ability. The results of the review highlight the heterogeneity of the methods used across manuscripts in terms of assessed clinical variables, imaging, and data analysis methods. This is particularly significant as, if the subjects' criteria are not carefully considered, results are easily prone to confounding factors.
Collapse
Affiliation(s)
- Jessica Rosenberg
- Institute of Neuroscience and Medicine (INM-4), Medical Imaging Physics, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany; JARA - Translational Brain Medicine & INM-11, RWTH Aachen University, 52074 Aachen, Germany; Department of Neurology, University Clinic Aachen, 52074 Aachen, Germany.
| | - Nazim Lechea
- Institute of Neuroscience and Medicine (INM-4), Medical Imaging Physics, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
| | - Gael N Pentang
- Institute of Neuroscience and Medicine (INM-4), Medical Imaging Physics, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
| | - Nadim J Shah
- Institute of Neuroscience and Medicine (INM-4), Medical Imaging Physics, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany; JARA - Translational Brain Medicine & INM-11, RWTH Aachen University, 52074 Aachen, Germany; Department of Neurology, University Clinic Aachen, 52074 Aachen, Germany; Department of Electrical and Computer Systems Engineering, and Monash Biomedical Imaging, School of Psychological Sciences, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
24
|
Sona C, Kumar A, Dogra S, Kumar BA, Umrao D, Yadav PN. Docosahexaenoic acid modulates brain-derived neurotrophic factor via GPR40 in the brain and alleviates diabesity-associated learning and memory deficits in mice. Neurobiol Dis 2018; 118:94-107. [DOI: 10.1016/j.nbd.2018.07.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 06/22/2018] [Accepted: 07/04/2018] [Indexed: 12/19/2022] Open
|
25
|
Insulin-signaling Pathway Regulates the Degradation of Amyloid β-protein via Astrocytes. Neuroscience 2018; 385:227-236. [PMID: 29932983 DOI: 10.1016/j.neuroscience.2018.06.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 06/09/2018] [Accepted: 06/11/2018] [Indexed: 01/06/2023]
Abstract
Alzheimer's disease (AD) has been considered as a metabolic dysfunction disease associated with impaired insulin signaling. Determining the mechanisms underlying insulin signaling dysfunction and resistance in AD will be important for its treatment. Impaired clearance of amyloid-β peptide (Aβ) significantly contributes to amyloid accumulation, which is typically observed in the brain of AD patients. Reduced expression of important Aβ-degrading enzymes in the brain, such as neprilysin (NEP) and insulin-degrading enzyme (IDE), can promote Aβ deposition in sporadic late-onset AD patients. Here, we investigated whether insulin regulates the degradation of Aβ by inducing expression of NEP and IDE in cultured astrocytes. Treatment of astrocytes with insulin significantly reduced cellular NEP levels, but increased IDE expression. The effects of insulin on the expression of NEP and IDE involved activation of an extracellular signal-regulated kinase (ERK)-mediated pathway. The reduction in cellular NEP levels was associated with NEP secretion into the culture medium, whereas IDE was increased in the cell membranes. Moreover, insulin-treated astrocytes significantly facilitated the degradation of exogenous Aβ within the culture medium. Interestingly, pretreatment of astrocytes with an ERK inhibitor prior to insulin exposure markedly inhibited insulin-induced degradation of Aβ. These results suggest that insulin exposure enhanced Aβ degradation via an increase in NEP secretion and IDE expression in astrocytes, via activation of the ERK-mediated pathway. The inhibition of insulin signaling pathways delayed Aβ degradation by attenuating alterations in NEP and IDE levels and competition with insulin and Aβ. Our results provide further insight into the pathological relevance of insulin resistance in AD development.
Collapse
|
26
|
Gault VA, Hölscher C. GLP-1 receptor agonists show neuroprotective effects in animal models of diabetes. Peptides 2018; 100:101-107. [PMID: 29412810 DOI: 10.1016/j.peptides.2017.11.017] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/24/2017] [Accepted: 11/24/2017] [Indexed: 12/18/2022]
Abstract
Enzyme-resistant receptor agonists of the incretin hormone glucagon-like peptide-1 (GLP-1) have shown positive therapeutic effects in people with type 2 diabetes mellitus (T2DM). T2DM has detrimental effects on brain function and impairment of cognition and memory formation has been described. One of the underlying mechanisms is most likely insulin de-sensitization in the brain, as insulin improves cognitive impairments and enhances learning. Treatment with GLP-1 receptor agonists improves memory formation and impairment of synaptic plasticity observed in animal models of diabetes-obesity. Furthermore, it has been shown that diabetes impairs growth factor signalling in the brain and reduces energy utilization in the cortex. Inflammation and apoptotic signalling was also increased. Treatment with GLP-1 receptor agonists improved neuronal growth and repair and reduced inflammation and apoptosis as well as oxidative stress. In comparison with the diabetes drug metformin, GLP-1 receptor agonists were able to improve glycemic control and reverse brain impairments, whereas metformin only normalized blood glucose levels. Clinical studies in non-diabetic patients with neurodegenerative disorders showed neuroprotective effects following administration with GLP-1 receptor agonists, demonstrating that neuroprotective effects are independent of blood glucose levels.
Collapse
Affiliation(s)
- Victor A Gault
- School of Biomedical Sciences, University of University, Coleraine, BT52 1SA, UK
| | - Christian Hölscher
- Biomedical and Life Sciences, Lancaster University, Lancaster, LA1 4YQ, UK.
| |
Collapse
|
27
|
Kharabian Masouleh S, Beyer F, Lampe L, Loeffler M, Luck T, Riedel-Heller SG, Schroeter ML, Stumvoll M, Villringer A, Witte AV. Gray matter structural networks are associated with cardiovascular risk factors in healthy older adults. J Cereb Blood Flow Metab 2018; 38:360-372. [PMID: 28857651 PMCID: PMC5951018 DOI: 10.1177/0271678x17729111] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
While recent 'big data' analyses discovered structural brain networks that alter with age and relate to cognitive decline, identifying modifiable factors that prevent these changes remains a major challenge. We therefore aimed to determine the effects of common cardiovascular risk factors on vulnerable gray matter (GM) networks in a large and well-characterized population-based cohort. In 616 healthy elderly (258 women, 60-80 years) of the LIFE-Adult-Study, we assessed the effects of obesity, smoking, blood pressure, markers of glucose and lipid metabolism as well as physical activity on major GM-networks derived using linked independent component analysis. Age, sex, hypertension, diabetes, white matter hyperintensities, education and depression were considered as confounders. Results showed that smoking, higher blood pressure, and higher glycated hemoglobin (HbA1c) were independently associated with lower GM volume and thickness in GM-networks that covered most areas of the neocortex. Higher waist-to-hip ratio was independently associated with lower GM volume in a network of multimodal regions that correlated negatively with age and memory performance. In this large cross-sectional study, we found selective negative associations of smoking, higher blood pressure, higher glucose, and visceral obesity with structural covariance networks, suggesting that reducing these factors could help to delay late-life trajectories of GM aging.
Collapse
Affiliation(s)
| | - Frauke Beyer
- 1 Department of Neurology, Max Planck Institute for Cognitive and Brain Sciences, Leipzig, Germany
| | - Leonie Lampe
- 1 Department of Neurology, Max Planck Institute for Cognitive and Brain Sciences, Leipzig, Germany.,2 LIFE - Leipzig Research Center for Civilization Diseases, 9180 University of Leipzig , Leipzig, Germany
| | - Markus Loeffler
- 2 LIFE - Leipzig Research Center for Civilization Diseases, 9180 University of Leipzig , Leipzig, Germany.,3 Institute for Medical Informatics, Statistics, and Epidemiology (IMISE), 9180 University of Leipzig , Leipzig, Germany
| | - Tobias Luck
- 2 LIFE - Leipzig Research Center for Civilization Diseases, 9180 University of Leipzig , Leipzig, Germany.,4 Institute of Social Medicine, Occupational Health and Public Health (ISAP), Medical Faculty, 9180 University of Leipzig , Leipzig, Germany
| | - Steffi G Riedel-Heller
- 4 Institute of Social Medicine, Occupational Health and Public Health (ISAP), Medical Faculty, 9180 University of Leipzig , Leipzig, Germany
| | - Matthias L Schroeter
- 1 Department of Neurology, Max Planck Institute for Cognitive and Brain Sciences, Leipzig, Germany.,2 LIFE - Leipzig Research Center for Civilization Diseases, 9180 University of Leipzig , Leipzig, Germany.,5 Clinic for Cognitive Neurology, 9180 University of Leipzig , Leipzig, Germany
| | - Michael Stumvoll
- 6 IFB Adiposity Diseases Faculty of Medicine, 9180 University of Leipzig , Leipzig, Germany
| | - Arno Villringer
- 1 Department of Neurology, Max Planck Institute for Cognitive and Brain Sciences, Leipzig, Germany.,5 Clinic for Cognitive Neurology, 9180 University of Leipzig , Leipzig, Germany
| | - A Veronica Witte
- 1 Department of Neurology, Max Planck Institute for Cognitive and Brain Sciences, Leipzig, Germany
| |
Collapse
|
28
|
Novel dual GLP-1/GIP receptor agonists show neuroprotective effects in Alzheimer's and Parkinson's disease models. Neuropharmacology 2018; 136:251-259. [PMID: 29402504 DOI: 10.1016/j.neuropharm.2018.01.040] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 01/10/2018] [Accepted: 01/27/2018] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes is a risk factor for several chronic neurodegenerative disorders such as Alzheimer's or Parkinson's disease. The link appears to be insulin de-sensitisation in the brain. Insulin is an important neuroprotective growth factor. GLP-1 and GIP are growth factors that re-sensitise insulin and GLP-1 mimetics are used in the clinic to treat diabetes. GLP-1 and GIP mimetics initially designed to treat diabetes show good protective effects in animal models of Alzheimer's and Parkinson's disease. Based on these results, several clinical trials have shown first encouraging effects in patients with Alzheimer's or Parkinson' disease. Novel dual GLP-1/GIP receptor agonists have been developed to treat diabetes, and they also show good neuroprotective effects that are superior to single GLP-1 analogues. Several newer dual analogues have been tested that have been engineered to cross the blood -brain barrier. They show clear neuroprotective effects by reducing inflammation and oxidative stress and apoptotic signalling and protecting memory formation, synaptic numbers and synaptic activity, motor activity, dopaminergic neurons, cortical activity and energy utilisation in the brain. These results demonstrate the potential of developing disease-modifying treatments for Alzheimer's and Parkinson's disease that are superior to current single GLP-1 mimetics. This article is part of the Special Issue entitled 'Metabolic Impairment as Risk Factors for Neurodegenerative Disorders.'
Collapse
|
29
|
Li W, Huang E. An Update on Type 2 Diabetes Mellitus as a Risk Factor for Dementia. J Alzheimers Dis 2018; 53:393-402. [PMID: 27163819 DOI: 10.3233/jad-160114] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
With the rapidly expanding evidence on brain structural and functional changes in type 2 diabetes mellitus (T2DM) patients, there is an increasing need to update our understanding on how T2DM associates with dementia as well as the underlying pathophysiological mechanisms. A literature search of T2DM and dementia or cognition impairments was carried out in electronic databases Medline, EMBASE, and Google Scholar. In this review, the chosen evidence was limited to human subject studies only, and data on either type 1 diabetes mellitus (T1DM) or non-classified diabetes were excluded. T2DM is a risk factor for both vascular dementia (VaD) and Alzheimer's disease (AD), although AD pathological marker studies have not provided sufficient evidence. T2DM interacts additively or synergistically with many factors, including old age, hypertension, total cholesterol, and APOEɛ4 carrier status for impaired cognition functions seen in patients with T2DM. In addition, comorbid T2DM can worsen the clinical presentations of patients with either AD or VaD. In summary, T2DM increases the risk for AD through different mechanisms for VaD although some mechanisms may overlap. Tau-related neurofibrillary tangles instead of amyloid-β plaques are more likely to be the pathological biomarkers for T2DM-related dementia. Degeneration of neurons in the brain, impaired regional blood supply/metabolism, and genetic predisposition are all involved in T2DM-associated dementia or cognitive impairments.
Collapse
Affiliation(s)
- Wei Li
- Master of Physician Assistant Studies, School of Health and Rehabilitation Sciences, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Edgar Huang
- School of Informatics and Computing, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| |
Collapse
|
30
|
The diabetic brain and cognition. J Neural Transm (Vienna) 2017; 124:1431-1454. [PMID: 28766040 DOI: 10.1007/s00702-017-1763-2] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/13/2017] [Indexed: 12/20/2022]
Abstract
The prevalence of both Alzheimer's disease (AD) and vascular dementia (VaD) is increasing with the aging of the population. Studies from the last several years have shown that people with diabetes have an increased risk for dementia and cognitive impairment. Therefore, the authors of this consensus review tried to elaborate on the role of diabetes, especially diabetes type 2 (T2DM) in both AD and VaD. Based on the clinical and experimental work of scientists from 18 countries participating in the International Congress on Vascular Disorders and on literature search using PUBMED, it can be concluded that T2DM is a risk factor for both, AD and VaD, based on a pathology of glucose utilization. This pathology is the consequence of a disturbance of insulin-related mechanisms leading to brain insulin resistance. Although the underlying pathological mechanisms for AD and VaD are different in many aspects, the contribution of T2DM and insulin resistant brain state (IRBS) to cerebrovascular disturbances in both disorders cannot be neglected. Therefore, early diagnosis of metabolic parameters including those relevant for T2DM is required. Moreover, it is possible that therapeutic options utilized today for diabetes treatment may also have an effect on the risk for dementia. T2DM/IRBS contribute to pathological processes in AD and VaD.
Collapse
|
31
|
Shafei MA, Harris M, Conway ME. Divergent Metabolic Regulation of Autophagy and mTORC1-Early Events in Alzheimer's Disease? Front Aging Neurosci 2017. [PMID: 28626421 PMCID: PMC5454035 DOI: 10.3389/fnagi.2017.00173] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive disease associated with the production and deposition of amyloid β-peptide (Aβ) aggregates and neurofibrillary tangles, which lead to synaptic and neuronal damage. Reduced autophagic flux has been widely associated with the accumulation of autophagic vacuoles (AV), which has been proposed to contribute to aggregate build-up observed in AD. As such, targeting autophagy regulation has received wide review, where an understanding as to how this mechanism can be controlled will be important to neuronal health. The mammalian target of rapamycin complex 1 (mTORC1), which was found to be hyperactive in AD brain, regulates autophagy and is considered to be mechanistically important to aberrant autophagy in AD. Hormones and nutrients such as insulin and leucine, respectively, positively regulate mTORC1 activation and are largely considered to inhibit autophagy. However, in AD brain there is a dysregulation of nutrient metabolism, linked to insulin resistance, where a role for insulin treatment to improve cognition has been proposed. Recent studies have highlighted that mitochondrial proteins such as glutamate dehydrogenase and the human branched chain aminotransferase protein, through metabolism of leucine and glutamate, differentially regulate mTORC1 and autophagy. As the levels of the hBCAT proteins are significantly increased in AD brain relative to aged-matched controls, we discuss how these metabolic pathways offer new potential therapeutic targets. In this review article, we highlight the core regulation of autophagy through mTORC1, focusing on how insulin and leucine will be important to consider in particular with respect to our understanding of nutrient load and AD pathogenesis.
Collapse
Affiliation(s)
- Mai A Shafei
- Department of Applied Science, The University of the West of EnglandBristol, United Kingdom
| | - Matthew Harris
- Department of Applied Science, The University of the West of EnglandBristol, United Kingdom
| | - Myra E Conway
- Department of Applied Science, The University of the West of EnglandBristol, United Kingdom
| |
Collapse
|
32
|
Beyer F, Kharabian Masouleh S, Huntenburg JM, Lampe L, Luck T, Riedel-Heller SG, Loeffler M, Schroeter ML, Stumvoll M, Villringer A, Witte AV. Higher body mass index is associated with reduced posterior default mode connectivity in older adults. Hum Brain Mapp 2017; 38:3502-3515. [PMID: 28397392 DOI: 10.1002/hbm.23605] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/24/2017] [Accepted: 03/27/2017] [Indexed: 12/31/2022] Open
Abstract
Obesity is a complex neurobehavioral disorder that has been linked to changes in brain structure and function. However, the impact of obesity on functional connectivity and cognition in aging humans is largely unknown. Therefore, the association of body mass index (BMI), resting-state network connectivity, and cognitive performance in 712 healthy, well-characterized older adults of the Leipzig Research Center for Civilization Diseases (LIFE) cohort (60-80 years old, mean BMI 27.6 kg/m2 ± 4.2 SD, main sample: n = 521, replication sample: n = 191) was determined. Statistical analyses included a multivariate model selection approach followed by univariate analyses to adjust for possible confounders. Results showed that a higher BMI was significantly associated with lower default mode functional connectivity in the posterior cingulate cortex and precuneus. The effect remained stable after controlling for age, sex, head motion, registration quality, cardiovascular, and genetic factors as well as in replication analyses. Lower functional connectivity in BMI-associated areas correlated with worse executive function. In addition, higher BMI correlated with stronger head motion. Using 3T neuroimaging in a large cohort of healthy older adults, independent negative associations of obesity and functional connectivity in the posterior default mode network were observed. In addition, a subtle link between lower resting-state connectivity in BMI-associated regions and cognitive function was found. The findings might indicate that obesity is associated with patterns of decreased default mode connectivity similar to those seen in populations at risk for Alzheimer's disease. Hum Brain Mapp 38:3502-3515, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Frauke Beyer
- Department of Neurology, Max Planck Institute for Cognitive and Brain Sciences, Leipzig, Germany.,Subproject A1, Collaborative Research Centre 1052 "Obesity Mechanisms", University of Leipzig, Leipzig, Germany
| | | | - Julia M Huntenburg
- Max Planck Research Group for Neuroanatomy and Connectivity, Max Planck Institute for Cognitive and Brain Sciences, Leipzig, Germany
| | - Leonie Lampe
- Department of Neurology, Max Planck Institute for Cognitive and Brain Sciences, Leipzig, Germany.,LIFE - Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Tobias Luck
- LIFE - Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany.,Institute of Social Medicine, Occupational Health and Public Health (ISAP), University of Leipzig, Leipzig, Germany
| | - Steffi G Riedel-Heller
- LIFE - Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany.,Institute of Social Medicine, Occupational Health and Public Health (ISAP), University of Leipzig, Leipzig, Germany
| | - Markus Loeffler
- LIFE - Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Matthias L Schroeter
- LIFE - Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany.,Department of Cognitive Neurology, University of Leipzig, Leipzig, Germany
| | - Michael Stumvoll
- Subproject A1, Collaborative Research Centre 1052 "Obesity Mechanisms", University of Leipzig, Leipzig, Germany.,IFB Adiposity Diseases Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Arno Villringer
- Department of Neurology, Max Planck Institute for Cognitive and Brain Sciences, Leipzig, Germany.,Subproject A1, Collaborative Research Centre 1052 "Obesity Mechanisms", University of Leipzig, Leipzig, Germany.,LIFE - Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - A Veronica Witte
- Department of Neurology, Max Planck Institute for Cognitive and Brain Sciences, Leipzig, Germany.,Subproject A1, Collaborative Research Centre 1052 "Obesity Mechanisms", University of Leipzig, Leipzig, Germany
| |
Collapse
|
33
|
Hölscher C. Glucagon-like peptide 1 and glucose-dependent insulinotropic polypeptide analogues as novel treatments for Alzheimer’s and Parkinson’s disease. Cardiovasc Endocrinol 2016. [DOI: 10.1097/xce.0000000000000087] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
34
|
Abstract
Painful neuropathy, like the other complications of diabetes, is a growing healthcare concern. Unfortunately, current treatments are of variable efficacy and do not target underlying pathogenic mechanisms, in part because these mechanisms are not well defined. Rat and mouse models of type 1 diabetes are frequently used to study diabetic neuropathy, with rats in particular being consistently reported to show allodynia and hyperalgesia. Models of type 2 diabetes are being used with increasing frequency, but the current literature on the progression of indices of neuropathic pain is variable and relatively few therapeutics have yet been developed in these models. While evidence for spontaneous pain in rodent models is sparse, measures of evoked mechanical, thermal and chemical pain can provide insight into the pathogenesis of the condition. The stocking and glove distribution of pain tantalizingly suggests that the generator site of neuropathic pain is found within the peripheral nervous system. However, emerging evidence demonstrates that amplification in the spinal cord, via spinal disinhibition and neuroinflammation, and also in the brain, via enhanced thalamic activity or decreased cortical inhibition, likely contribute to the pathogenesis of painful diabetic neuropathy. Several potential therapeutic strategies have emerged from preclinical studies, including prophylactic treatments that intervene against underlying mechanisms of disease, treatments that prevent gains of nociceptive function, treatments that suppress enhancements of nociceptive function, and treatments that impede normal nociceptive mechanisms. Ongoing challenges include unraveling the complexity of underlying pathogenic mechanisms, addressing the potential disconnect between the perceived location of pain and the actual pain generator and amplifier sites, and finding ways to identify which mechanisms operate in specific patients to allow rational and individualized choice of targeted therapies.
Collapse
Affiliation(s)
- Corinne A Lee-Kubli
- Graduate School of Biomedical Sciences, Sanford-Burnham Institute for Molecular Medicine, La Jolla, CA, USA; Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Nigel A Calcutt
- Department of Pathology, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
35
|
Abstract
Although an association between diabetes mellitus (DM) and cognitive dysfunction has been recognized for a century, it is often not considered as a complication of DM and remains under-recognized. Cognitive dysfunction, usually present as mild cognitive impairment, can occur with either type 1 or type 2 DM. Both forms of DM contribute to accelerated cerebral atrophy and to the presence of heightened white matter abnormalities. These effects are noted most at the two extremes of life, in childhood and in the advanced years. The cognitive spheres most affected include attention and executive function, processing speed, perception, and memory. Although DM is unlikely to lead to frank dementia, its ability to exacerbate existing neurodegenerative processes, such as Alzheimer disease, will impact tremendously upon our society in the upcoming decades as our population ages. This chapter describes the clinical impact of DM upon the brain, along with discussion of the potential therapeutic avenues to be discovered in the coming decades. We need to prepare for better preventative and therapeutic management of this cerebral neurodegenerative condition.
Collapse
Affiliation(s)
- Cory Toth
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
36
|
Johnson LA, Gamboa A, Vintimilla R, Cheatwood AJ, Grant A, Trivedi A, Edwards M, Hall JR, O'Bryant SE. Comorbid Depression and Diabetes as a Risk for Mild Cognitive Impairment and Alzheimer's Disease in Elderly Mexican Americans. J Alzheimers Dis 2016; 47:129-36. [PMID: 26402761 DOI: 10.3233/jad-142907] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND The links between diabetes, depression, and Alzheimer's disease (AD) has been established, but they are still poorly understood. However, little research has examined the effect that comorbidity of depression and diabetes has on cognitive impairment in an ethnically diverse sample. OBJECTIVE The purpose of this study was to investigate the relationship between comorbid diabetes and depression on cognitive dysfunction; and examine the relationship in an ethnically diverse population. METHODS AND RESULTS Analyses of data from 2,436 participants (914 men and 1,522 women) of three separate cohorts: HABLE, FRONTIER, and TARCC. In the HABLE cohort, comorbidity (odds ratio [OR] = 3.008; 95% CI = 1.358-6.667), age (OR = 1.138; 95% CI = 1.093-1.185), and education (OR = 0.915; 95% CI = 0.852-0.982) increased the risk of mild cognitive impairment (MCI) diagnosis among elderly Mexican American. In the TARCC cohort, results showed an increase risk of MCI in both non-Hispanic whites (OR = 18.795; 95% CI = 2.229-158.485) and Mexican Americans (OR = 8.417; 95% CI = 2.967-23.878). Finally, results in the FRONTIER cohort showed that in elderly Mexican Americans, comorbidity (OR = 2.754; 95% CI = 1.084-6.995) and age (OR = 1.069; 95% CI = 1.023-1.118) significantly increased risk of MCI. In non-Hispanic whites, comorbidity did not significantly increase risk of MCI. CONCLUSIONS Among elderly Mexican Americans, comorbid depression and diabetes significantly increased risk for MCI and AD across cohorts. Effects of comorbid diabetes and depression on MCI were inconclusive. Our results support the link between comorbid diabetes and depression and risk for cognitive decline among Mexican Americans. This finding is of critical importance as the Hispanic population is at higher risk of developing AD.
Collapse
Affiliation(s)
- Leigh A Johnson
- Department of Internal Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA.,Institute for Aging & Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Adriana Gamboa
- Department of Internal Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Raul Vintimilla
- Department of Internal Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | | | | | | | - Melissa Edwards
- Department of Internal Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - James R Hall
- Institute for Aging & Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX, USA.,Department of Psychiatry, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Sid E O'Bryant
- Department of Internal Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA.,Institute for Aging & Alzheimer's Disease Research, University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
37
|
Diabetes, Gray Matter Loss, and Cognition in the Setting of Parkinson Disease. Acad Radiol 2016; 23:577-81. [PMID: 26874576 DOI: 10.1016/j.acra.2015.07.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 06/25/2015] [Accepted: 07/21/2015] [Indexed: 11/21/2022]
Abstract
RATIONALE AND OBJECTIVES Parkinson disease (PD) is a progressive neurodegenerative disorder affecting motor and cognitive functions. Prior studies showed that patients with PD and diabetes (DM) demonstrate worse clinical outcomes compared to nondiabetic subjects with PD. Our study aimed at defining the relationship between DM, gray matter volume, and cognition in patients with PD. MATERIALS AND METHODS This study included 36 subjects with PD (12 with DM, 24 without DM, mean age = 66). Subjects underwent high-resolution T1-weighted brain magnetic resonance imaging, [(11)C]dihydrotetrabenazine positron emission tomography imaging to quantify nigrostriatal dopaminergic denervation, clinical, and cognitive assessments. Magnetic resonance images were postprocessed to determine total and lobar cortical gray matter volumes. Cognitive testing scores were converted to z-scores for specific cognitive domains and a composite global cognitive z-score based on normative data computed. Analysis of covariance, accounting for effects of age, gender, intracranial volume, and striatal [(11)C]dihydrotetrabenazine binding, was used to test the relationship between DM and gray matter volumes. RESULTS Impact of DM on total gray matter volume was significant (P = 0.02). Post hoc analyses of lobar cortical gray matter volumes revealed that DM was more selectively associated with lower gray matter volumes in the frontal regions (P = 0.01). Cognitive post hoc analyses showed that interaction of total gray matter volume and DM status was significantly associated with composite (P = 0.007), executive (P = 0.02), and visuospatial domain cognitive z-scores (P = 0.005). These associations were also significant for the frontal cortical gray matter. CONCLUSION DM may exacerbate brain atrophy and cognitive functions in PD with greater vulnerability in the frontal lobes. Given the high prevalence of DM in the elderly, delineating its effects on patient outcomes in the PD population is of importance.
Collapse
|
38
|
Kharabian Masouleh S, Arélin K, Horstmann A, Lampe L, Kipping JA, Luck T, Riedel-Heller SG, Schroeter ML, Stumvoll M, Villringer A, Witte AV. Higher body mass index in older adults is associated with lower gray matter volume: implications for memory performance. Neurobiol Aging 2016; 40:1-10. [DOI: 10.1016/j.neurobiolaging.2015.12.020] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 12/28/2015] [Accepted: 12/28/2015] [Indexed: 10/22/2022]
|
39
|
McGregor G, Malekizadeh Y, Harvey J. Minireview: Food for thought: regulation of synaptic function by metabolic hormones. Mol Endocrinol 2016; 29:3-13. [PMID: 25470238 DOI: 10.1210/me.2014-1328] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The peripheral actions of the metabolic hormones, leptin and insulin, are well documented. However, the functions of these hormones are not restricted to the periphery because evidence is growing that both leptin and insulin can readily cross the blood-brain barrier and have widespread central actions. The hippocampus in particular expresses high levels of both insulin and leptin receptors as well as key components of their associated signaling cascades. Moreover, recent studies indicate that both hormones are potential cognitive enhancers. Indeed, it has been demonstrated that both leptin and insulin markedly influence key cellular events that underlie hippocampal learning and memory including activity-dependent synaptic plasticity and the trafficking of glutamate receptors to and away from hippocampal synapses. The hippocampal formation is also a prime site for the neurodegenerative processes that occur during Alzheimer's disease, and impairments in either leptin or insulin function have been linked to central nervous system-driven diseases like Alzheimer's disease. Thus, the capacity of the metabolic hormones, leptin and insulin, to regulate hippocampal synaptic function has significant implications for normal brain function and also central nervous system-driven disease.
Collapse
Affiliation(s)
- Gemma McGregor
- Division of Neuroscience, Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, United Kingdom
| | | | | |
Collapse
|
40
|
Vicente Miranda H, El-Agnaf OMA, Outeiro TF. Glycation in Parkinson's disease and Alzheimer's disease. Mov Disord 2016; 31:782-90. [PMID: 26946341 DOI: 10.1002/mds.26566] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 12/21/2015] [Accepted: 01/07/2016] [Indexed: 12/14/2022] Open
Abstract
Glycation is a spontaneous age-dependent posttranslational modification that can impact the structure and function of several proteins. Interestingly, glycation can be detected at the periphery of Lewy bodies in the brain in Parkinson's disease. Moreover, α-synuclein can be glycated, at least under experimental conditions. In Alzheimer's disease, glycation of amyloid β peptide exacerbates its toxicity and contributes to neurodegeneration. Recent studies establish diabetes mellitus as a risk factor for several neurodegenerative disorders, including Parkinson's and Alzheimer's diseases. However, the mechanisms underlying this connection remain unclear. We hypothesize that hyperglycemia might play an important role in the development of these disorders, possibly by also inducing protein glycation and thereby dysfunction, aggregation, and deposition. Here, we explore protein glycation as a common player in Parkinson's and Alzheimer's diseases and propose it may constitute a novel target for the development of strategies for neuroprotective therapeutic interventions. © 2016 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Omar M A El-Agnaf
- Neurological Disorders Center, Qatar Biomedical Research Institute, and College of Science and Engineering, Hamad Bin Khalifa University (HBKU), Education City, Qatar Foundation, P.O. Box 5825 Doha, Qatar
| | - Tiago Fleming Outeiro
- CEDOC - Chronic Diseases Research Center, NOVA Medical School, Lisboa, Portugal.,Department of Neurodegeneration and Restorative Research, Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University Medical Center Goettingen, Goettingen, Germany.,Max Planck Institute for Experimental Medicine, Goettingen, Germany
| |
Collapse
|
41
|
Park SE, Kim H, Lee J, Lee NK, Hwang JW, Yang JJ, Ye BS, Cho H, Kim HJ, Kim YJ, Jung NY, Son TO, Cho EB, Jang H, Jang EY, Hong CH, Lee JM, Kang M, Shin HY, Na DL, Seo SW. Decreased hemoglobin levels, cerebral small-vessel disease, and cortical atrophy: among cognitively normal elderly women and men. Int Psychogeriatr 2016; 28:147-56. [PMID: 25990664 DOI: 10.1017/s1041610215000733] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Decreased hemoglobin levels increase the risk of developing dementia among the elderly. However, the underlying mechanisms that link decreased hemoglobin levels to incident dementia still remain unclear, possibly due to the fact that few studies have reported on the relationship between low hemoglobin levels and neuroimaging markers. We, therefore, investigated the relationships between decreased hemoglobin levels, cerebral small-vessel disease (CSVD), and cortical atrophy in cognitively healthy women and men. METHODS Cognitively normal women (n = 1,022) and men (n = 1,018) who underwent medical check-ups and magnetic resonance imaging (MRI) were enrolled at a health promotion center. We measured hemoglobin levels, white matter hyperintensities (WMH) scales, lacunes, and microbleeds. Cortical thickness was automatically measured using surface based methods. Multivariate regression analyses were performed after controlling for possible confounders. RESULTS Decreased hemoglobin levels were not associated with the presence of WMH, lacunes, or microbleeds in women and men. Among women, decreased hemoglobin levels were associated with decreased cortical thickness in the frontal (Estimates, 95% confidence interval, -0.007, (-0.013, -0.001)), temporal (-0.010, (-0.018, -0.002)), parietal (-0.009, (-0.015, -0.003)), and occipital regions (-0.011, (-0.019, -0.003)). Among men, however, no associations were observed between hemoglobin levels and cortical thickness. CONCLUSION Our findings suggested that decreased hemoglobin levels affected cortical atrophy, but not increased CSVD, among women, although the association is modest. Given the paucity of modifiable risk factors for age-related cognitive decline, our results have important public health implications.
Collapse
Affiliation(s)
- Sang Eon Park
- Department of Neurology,Samsung Medical Center,Sungkyunkwan University School of Medicine,Seoul,South Korea
| | - Hojeong Kim
- Department of Neurology,Samsung Medical Center,Sungkyunkwan University School of Medicine,Seoul,South Korea
| | - Jeongmin Lee
- Department of Neurology,Samsung Medical Center,Sungkyunkwan University School of Medicine,Seoul,South Korea
| | - Na Kyung Lee
- Department of Neurology,Samsung Medical Center,Sungkyunkwan University School of Medicine,Seoul,South Korea
| | - Jung Won Hwang
- Department of Neurology,Samsung Medical Center,Sungkyunkwan University School of Medicine,Seoul,South Korea
| | - Jin-ju Yang
- Department of Biomedical Engineering,Hanyang University,Seoul,South Korea
| | - Byoung Seok Ye
- Department of Neurology,Yonsei University College of Medicine,Seoul,South Korea
| | - Hanna Cho
- Department of Neurology,Gangnam Severance Hospital,Yonsei University College of Medicine,Seoul,South Korea
| | - Hee Jin Kim
- Department of Neurology,Samsung Medical Center,Sungkyunkwan University School of Medicine,Seoul,South Korea
| | - Yeo Jin Kim
- Department of Neurology,Samsung Medical Center,Sungkyunkwan University School of Medicine,Seoul,South Korea
| | - Na-Yeon Jung
- Department of Neurology,Samsung Medical Center,Sungkyunkwan University School of Medicine,Seoul,South Korea
| | - Tae Ok Son
- Department of Neurology,Samsung Medical Center,Sungkyunkwan University School of Medicine,Seoul,South Korea
| | - Eun Bin Cho
- Department of Neurology,Samsung Medical Center,Sungkyunkwan University School of Medicine,Seoul,South Korea
| | - Hyemin Jang
- Department of Neurology,Samsung Medical Center,Sungkyunkwan University School of Medicine,Seoul,South Korea
| | - Eun Young Jang
- Department of Neurology,Samsung Medical Center,Sungkyunkwan University School of Medicine,Seoul,South Korea
| | | | - Jong-Min Lee
- Department of Biomedical Engineering,Hanyang University,Seoul,South Korea
| | - Mira Kang
- Center for Health Promotion,Samsung Medical Center,Seoul,South Korea
| | - Hee-Young Shin
- Center for Health Promotion,Samsung Medical Center,Seoul,South Korea
| | - Duk L Na
- Department of Neurology,Samsung Medical Center,Sungkyunkwan University School of Medicine,Seoul,South Korea
| | - Sang Won Seo
- Department of Neurology,Samsung Medical Center,Sungkyunkwan University School of Medicine,Seoul,South Korea
| |
Collapse
|
42
|
Quantitative validation of a visual rating scale for frontal atrophy: associations with clinical status, APOE e4, CSF biomarkers and cognition. Eur Radiol 2015; 26:2597-610. [PMID: 26560730 DOI: 10.1007/s00330-015-4101-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 10/14/2015] [Accepted: 10/30/2015] [Indexed: 10/22/2022]
Abstract
OBJECTIVES To validate a visual rating scale of frontal atrophy with quantitative imaging and study its association with clinical status, APOE ε4, CSF biomarkers, and cognition. METHODS The AddNeuroMed and ADNI cohorts were combined giving a total of 329 healthy controls, 421 mild cognitive impairment patients, and 286 Alzheimer's disease (AD) patients. Thirty-four patients with frontotemporal dementia (FTD) were also included. Frontal atrophy was assessed with the frontal sub-scale of the global cortical atrophy scale (GCA-F) on T1-weighted images. Automated imaging markers of cortical volume, thickness, and surface area were evaluated. Manual tracing was also performed. RESULTS The GCA-F scale reliably reflects frontal atrophy, with orbitofrontal, dorsolateral, and motor cortices being the regions contributing most to the GCA-F ratings. GCA-F primarily reflects reductions in cortical volume and thickness, although it was able to detect reductions in surface area too. The scale showed significant associations with clinical status and cognition. CONCLUSION The GCA-F scale may have implications for clinical practice as supportive diagnostic tool for disorders demonstrating predominant frontal atrophy such as FTD and the executive presentation of AD. We believe that GCA-F is feasible for use in clinical routine for the radiological assessment of dementia and other disorders. KEY POINTS • The GCA-F visual rating scale reliably reflects frontal brain atrophy. • Orbitofrontal, dorsolateral, and motor cortices are the most contributing regions. • GCA-F shows significant associations with clinical status and cognition. • GCA-F may be supportive diagnostic tool for disorders demonstrating predominant frontal atrophy. • GCA-F may be feasible for use in radiological routine.
Collapse
|
43
|
Del Bene A, Ciolli L, Borgheresi L, Poggesi A, Inzitari D, Pantoni L. Is type 2 diabetes related to leukoaraiosis? an updated review. Acta Neurol Scand 2015; 132:147-55. [PMID: 25772411 DOI: 10.1111/ane.12398] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2015] [Indexed: 02/04/2023]
Abstract
A significantly increased interest has been dedicated to the study of the effects of diabetes mellitus (DM) on the brain. DM is associated with an increased risk of stroke and cognitive decline. In patients with DM, neuroimaging discloses with high-frequency structural changes, such as cerebral atrophy, infarcts and white matter lesions, also called leukoaraiosis (LA), an expression of small vessel disease. A previous review showed a relation between DM and both cerebral atrophy and lacunar infarcts, while the question about the relation between DM and LA remained unanswered. In this review, we provide an update on data on this last association. In the reviewed studies, we examined the presence of DM, other disease characteristics, such as duration and complications, and laboratory markers of the disease such as blood glycated hemoglobin (HbA1c), insulin resistance, insulin concentrations and their association with LA. About 40% of the reviewed studies reported a statistically significant association between DM and LA. Long-standing DM and a poor glycemic control were associated with severe LA. Studies using innovative MRI techniques, such as diffusion tensor imaging (DTI), reported a significant association between microstructural white matter alterations and DM. This review highlights more firmly than previously reported the existence of a relation between DM and both presence and severity of LA. These results are possibly due to more sensitive and advanced imaging techniques recently used to study the extent of LA. However, because of the heterogeneous methodology used in the reviewed studies, a definitive conclusion cannot be drawn.
Collapse
Affiliation(s)
- A. Del Bene
- NEUROFARBA Department; Neuroscience Section; University of Florence; Florence Italy
| | - L. Ciolli
- NEUROFARBA Department; Neuroscience Section; University of Florence; Florence Italy
| | - L. Borgheresi
- NEUROFARBA Department; Neuroscience Section; University of Florence; Florence Italy
| | - A. Poggesi
- NEUROFARBA Department; Neuroscience Section; University of Florence; Florence Italy
| | - D. Inzitari
- NEUROFARBA Department; Neuroscience Section; University of Florence; Florence Italy
| | - L. Pantoni
- Stroke Unit and Neurology; Azienda Ospedaliero Universitaria Careggi; Florence Italy
| |
Collapse
|
44
|
Ferreira D, Cavallin L, Larsson EM, Muehlboeck JS, Mecocci P, Vellas B, Tsolaki M, Kłoszewska I, Soininen H, Lovestone S, Simmons A, Wahlund LO, Westman E. Practical cut-offs for visual rating scales of medial temporal, frontal and posterior atrophy in Alzheimer's disease and mild cognitive impairment. J Intern Med 2015; 278:277-90. [PMID: 25752192 DOI: 10.1111/joim.12358] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Atrophy in the medial temporal lobe, frontal lobe and posterior cortex can be measured with visual rating scales such as the medial temporal atrophy (MTA), global cortical atrophy - frontal subscale (GCA-F) and posterior atrophy (PA) scales, respectively. However, practical cut-offs are urgently needed, especially now that different presentations of Alzheimer's disease (AD) are included in the revised diagnostic criteria. AIMS The aim of this study was to generate a list of practical cut-offs for the MTA, GCA-F and PA scales, for both diagnosis of AD and determining prognosis in mild cognitive impairment (MCI), and to evaluate the influence of key demographic and clinical factors on these cut-offs. METHODS AddNeuroMed and ADNI cohorts were combined giving a total of 1147 participants (322 patients with AD, 480 patients with MCI and 345 control subjects). The MTA, GCA-F and PA scales were applied and a broad range of cut-offs was evaluated. RESULTS The MTA scale showed better diagnostic and predictive performances than the GCA-F and PA scales. Age, apolipoprotein E (ApoE) ε4 status and age at disease onset influenced all three scales. For the age ranges 45-64, 65-74, 75-84 and 85-94 years, the following cut-offs should be used. MTA: ≥1.5, ≥1.5, ≥2 and ≥2.5; GCA-F, ≥1, ≥1, ≥1 and ≥1; and PA, ≥1, ≥1, ≥1 and ≥1, respectively, with an adjustment for early-onset ApoE ε4 noncarrier AD patients (MTA: ≥2, ≥2, ≥3 and ≥3; and GCA-F: ≥1, ≥1, ≥2 and ≥2, respectively). CONCLUSIONS If successfully validated in clinical settings, the list of practical cut-offs proposed here might be useful in clinical practice. Their use might also (i) promote research on atrophy subtypes, (ii) increase the understanding of different presentations of AD, (iii) improve diagnosis and prognosis and (iv) aid population selection and enrichment for clinical trials.
Collapse
Affiliation(s)
- D Ferreira
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Stockholm, Sweden
| | - L Cavallin
- Department of Clinical Science, Intervention and Technology, Division of Medical Imaging and Technology, Karolinska Institutet, Stockholm, Sweden.,Department of Radiology, Karolinska University Hospital, Stockholm, Sweden
| | - E-M Larsson
- Department of Radiology, Oncology and Radiation Science, Uppsala University, Uppsala, Sweden
| | - J-S Muehlboeck
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Stockholm, Sweden
| | - P Mecocci
- Institute of Gerontology and Geriatrics, University of Perugia, Perugia, Italy
| | - B Vellas
- INSERM U 558, University of Toulouse, Toulouse, France
| | - M Tsolaki
- 3rd Department of Neurology, Aristoteleion Panepistimeion Thessalonikis, Thessaloniki, Greece
| | | | - H Soininen
- University of Eastern Finland, University Hospital of Kuopio, Kuopio, Finland
| | - S Lovestone
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, UK
| | - A Simmons
- Institute of Psychiatry, King's College London, London, UK.,NIHR Biomedical Research Centre for Mental Health, London, UK.,NIHR Biomedical Research Unit for Dementia, London, UK
| | - L-O Wahlund
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Stockholm, Sweden
| | - E Westman
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Stockholm, Sweden
| | | |
Collapse
|
45
|
Christensen A, Pike CJ. Menopause, obesity and inflammation: interactive risk factors for Alzheimer's disease. Front Aging Neurosci 2015. [PMID: 26217222 PMCID: PMC4493396 DOI: 10.3389/fnagi.2015.00130] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Alzheimer’s disease (AD) is a multifactorial neurodegenerative disorder, the development of which is regulated by several environmental and genetic risk factors. Two factors theorized to contribute to the initiation and/or progression of AD pathogenesis are age-related increases in inflammation and obesity. These factors may be particularly problematic in women. The onset of menopause in mid-life elevates the vulnerability of women to AD, an increased risk that is likely associated with the depletion of estrogens. Menopause is also linked with an abundance of additional changes, including increased central adiposity and inflammation. Here, we review the current literature to explore the interactions between obesity, inflammation, menopause and AD.
Collapse
Affiliation(s)
- Amy Christensen
- Davis School of Gerontology, University of Southern California Los Angeles, CA, USA
| | - Christian J Pike
- Davis School of Gerontology, University of Southern California Los Angeles, CA, USA
| |
Collapse
|
46
|
Hölscher C. First clinical data of the neuroprotective effects of nasal insulin application in patients with Alzheimer's disease. Alzheimers Dement 2014; 10:S33-7. [PMID: 24529523 DOI: 10.1016/j.jalz.2013.12.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 12/05/2013] [Indexed: 01/27/2023]
Abstract
Previous reviews have outlined the important role of insulin in the brain, and the observation that insulin signaling is desensitized in patients with Alzheimer's disease (AD). Because insulin is used to treat diabetes and insulin desensitization in the periphery, this motivated the design and execution of clinical pilot trials in patients with AD and mild cognitive impairment. Because insulin has powerful effects on blood sugar levels, a new technique was used by which insulin is applied as a spray. This method avoids high levels of insulin in the periphery and makes use of the transport system, via the nasal epithelium, into the brain. First trials in healthy subjects showed improvement in attention and memory tasks, and confirmed the concept that insulin signaling plays an important role in neuronal function and cognition. In a series of small clinical trials in patients with mild cognitive impairment/AD, nasal application of insulin or long-lasting insulin analogs showed improvements in memory tasks, cerebrospinal fluid biomarkers, and in a fluorodeoxyglucose positron emission tomographic study. In a more recent trial, two patient subgroups were identified, in which the insulin-resistant group improved after drug treatment whereas a subgroup that did not show insulin desensitization deteriorated. This highlights the need to conduct additional studies and demonstrates clearly that the hypothesis that insulin signaling plays in important role in cognition and AD has merit, and that this is a worthwhile target that shows great promise for future drug developments that improve insulin signaling. Insulin itself may not be the best choice, and other drugs that have been developed to treat diabetes that do not enhance insulin desensitization may be a better choice.
Collapse
Affiliation(s)
- Christian Hölscher
- Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, UK.
| |
Collapse
|
47
|
Romain G, Opacka-Juffry J. Cerebral ageing-the role of insulin and insulin-like growth factor signalling: A review. World J Neurol 2014; 4:12-22. [DOI: 10.5316/wjn.v4.i3.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 11/11/2014] [Accepted: 12/01/2014] [Indexed: 02/07/2023] Open
Abstract
Cerebral ageing is a complex biological process associated with progressing cerebrovascular disease and neuronal death. It does not always, however, associate with a functional decline, as the ageing mammalian brain retains considerable functional plasticity which supports successful cerebral ageing where age-related cognitive decline is modest. On the contrary, pathological cerebral ageing results in memory impairment and cognitive deterioration, with Alzheimer’s disease (AD) being a florid example. Trophic/growth factors promote brain plasticity; among them are peptides which belong to the insulin family. Preclinical research suggests that the evolutionarily conserved brain insulin/insulin-like growth factor-1 (IGF-1) signalling system controls lifespan and protects against some features of AD such as neurodegeneration-related accumulation of toxic proteins and cognitive deficiencies, as observed in animal models. Insulin and IGF-1 activate cell signalling mechanisms which play protective and regenerative roles; abnormalities in the insulin/IGF-1 system may trigger a cascade of neurodegeneration in AD. AD patients show cerebral resistance to insulin which associates with IGF-I resistance and dysregulation of insulin/IGF-1 receptors as well as cognitive deterioration. This review is focused on the roles of the insulin/IGF-1 signalling system in cerebral ageing and its potential involvement in neurodegeneration in the human brain as seen against the background of preclinical evidence.
Collapse
|
48
|
Takeuchi A, Matsushima E, Kato M, Konishi M, Izumiyama H, Murata Y, Hirata Y. Characteristics of neuropsychological functions in inpatients with poorly-controlled type 2 diabetes mellitus. J Diabetes Investig 2014; 3:325-30. [PMID: 24843583 PMCID: PMC4014957 DOI: 10.1111/j.2040-1124.2011.00170.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
UNLABELLED Aims/Introduction: It has been suggested that type 2 diabetes is associated with cognitive impairment. We investigated the neuropsychological profile of inpatients with poorly controlled type 2 diabetes and assessed the effects of clinical factors on neuropsychological functions. MATERIALS AND METHODS Forty-two patients with type 2 diabetes and 32 non diabetic control subjects were matched for age, sex ratio, and level of education. Attention & working memory, processing speed, verbal memory, visuospatial memory, visuoconstruction, and executive function were tested. Information about physical function, alcohol use, hypertension, dyslipidemia, and myocardial infarction was retrieved from personal interviews and medical records. RESULTS Diabetic patients demonstrated mild cognitive deterioration in attention & working memory, processing speed, verbal memory, and executive function. In particular, neuropsychological decline became prominent when tasks related with speed and verbal stimuli became unstructured and complex. Age was significantly associated with the majority of neuropsychological tests, whereas tasks dealing with working memory and executive function were associated with age only in the diabetic group. Duration of diabetes was associated with Backward Digit Span. CONCLUSIONS Accelerated aging had a major influence on cognitive decline in the diabetic group, whereas diminished performance in working memory and executive function might have been more related to diabetes-related cognitive impairment. (J Diabetes Invest, doi: 10.1111/j.2040-1124.2011.00170.x, 2011).
Collapse
Affiliation(s)
- Ai Takeuchi
- Section of Liaison Psychiatry & Palliative Medicine
| | | | - Motoichiro Kato
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo
| | - Mika Konishi
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo
| | - Hajime Izumiyama
- Clinical and Molecular Endocrinology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | | | - Yukio Hirata
- Clinical and Molecular Endocrinology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| |
Collapse
|
49
|
Abstract
The incretin hormone glucagon-like peptide 1 (GLP-1) has many effects in the body. It is best known for the 'incretin effect', facilitating insulin release from the pancreas under hyperglycaemic conditions. Building on this, GLP-1 mimetics have been developed as a treatment for type 2 diabetes. In the course of monitoring of patients, it has become apparent that GLP-1 mimetics have a range of other physiological effects in the body. In preclinical trials, a substantial body of evidence has been built that these mimetics have neuroprotective and anti-inflammatory effects. GLP-1 also has very similar growth-factor-like properties to insulin, which is presumably the underlying basis of the neuroprotective effects. In preclinical studies of Alzheimer's disease (AD), Parkinson's disease (PD), stroke and other neurodegenerative disorders, it has been shown that most GLP-1 mimetics cross the blood-brain barrier and show impressive neuroprotective effects in numerous studies. In animal models of AD, GLP-1 mimetics such as exendin-4, liraglutide and lixisenatide have shown protective effects in the CNS by reducing β-amyloid plaques, preventing loss of synapses and memory impairments, and reducing oxidative stress and the chronic inflammatory response in the brain. In animal models of PD, exendin-4 showed protection of dopaminergic neurons in the substantia nigra and prevention of dopamine loss in the basal ganglia while preserving motor control. These encouraging findings have spawned several clinical trials, some of which have shown encouraging initial results. Therefore, GLP-1 mimetics show great promise as a novel treatment for neurodegenerative conditions.
Collapse
Affiliation(s)
- Christian Hölscher
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YQ, UK
| |
Collapse
|
50
|
Insulin, incretins and other growth factors as potential novel treatments for Alzheimer's and Parkinson's diseases. Biochem Soc Trans 2014; 42:593-9. [DOI: 10.1042/bst20140016] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Recently, it has been shown that in patients with AD (Alzheimer's disease) and, to some degree, in patients with PD (Parkinson's disease) insulin signalling is impaired. This finding has initiated a range of research projects that showed remarkable improvements using treatments that initially had been developed to treat diabetes. Pre-clinical studies showed good neuroprotective effects when applying insulin or long-lasting analogues of incretin peptides. In transgenic animal models of AD and PD, analogues of the incretin GLP-1 (glucagon-like peptide 1) prevented neurodegenerative processes and improved neuronal and synaptic functionality in AD and PD. Amyloid plaque load and synaptic loss as well as cognitive impairment had been ameliorated in AD models, and dopaminergic loss of transmission and motor function was reversed in models of PD. On the basis of these promising findings, several clinical trials are being conducted with the first encouraging clinical results being published. In several pilot studies in AD patients, the nasal application of insulin showed encouraging effects on cognition and biomarkers. A pilot study in PD patients testing a GLP-1 receptor agonist that is currently on the market as a treatment for Type 2 diabetes also showed encouraging effects. Several other clinical trials are currently ongoing in AD patients. The present review summarizes the range of neuroprotective effects that these drugs have demonstrated and emphasizes the great promise that this approach has in providing novel treatments that have protective and even restorative properties that no current drug treatment can offer.
Collapse
|