1
|
Preechasuk L, Rilstone S, Tang WX, Man J, Yang M, Zhao E, Hoque L, Tuncay E, Wilding P, Godsland I, Halse O, Banerjee S, Oliver N, Hill NE. Glycaemic level and glycaemic variability in acute ischaemic stroke and functional outcome: An observational continuous glucose monitoring study. PLoS One 2025; 20:e0318456. [PMID: 40445939 PMCID: PMC12124563 DOI: 10.1371/journal.pone.0318456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 01/15/2025] [Indexed: 06/02/2025] Open
Abstract
INTRODUCTION Glycaemic variability has been associated with poor outcomes in critically ill patients. We aimed to study the association between glycaemic variability and functional outcome in patients with acute ischaemic stroke using continuous glucose monitoring to ensure all episodes of hyper- and hypoglycaemia were captured. RESEARCH DESIGN AND METHODS Participants with acute ischaemic stroke were enrolled and started blinded continuous glucose monitoring (Dexcom G6) between November 2020 and December 2022. Glucose data from the first 72 hours after admission were analysed. Patients were classified into 3 groups based on change in functional status (Modified Rankin Scale) between admission and discharge. These included (i) remained independent (RI); (ii) deteriorated to dependent (DD); and (iii) remained dependent (RD). RESULTS Data of 67 patients (mean±SD age 72.1 ± 14 years) were analysed; 19 participants had diabetes. The median (IQR) National Institutes of Health Stroke Scale (NIHSS) was 8 (3,14), and 34.3% received reperfusion therapy. The percentage of patients with RI, DD, and RD was 25.4, 55.2, 19.4%. Patients with DD had older age, higher rate of atrial fibrillation, systolic blood pressure, rate of in-hospital infection, NIHSS at admission and at 24 hours after reperfusion therapy compared to those RI. Continuous glucose monitoring was started at 38.4 (29.5,51) hours after stroke onset. Those with DD had higher mean glucose, %time above 180 mg/dL, and glucose standard deviation than the RI group at discharge. Multivariate analysis showed only an association between NIHSS at admission and deterioration in functional status. CONCLUSIONS In this pilot study, an association between glycaemic variability and functional deterioration after acute ischaemic stroke was not observed. Clinical Trial Registration numberNCT04521634.
Collapse
Affiliation(s)
- Lukana Preechasuk
- Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London, United Kingdom
- Siriraj Diabetes Center of Excellence, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok Noi, Bangkok, Thailand
| | - Siân Rilstone
- Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London, United Kingdom
| | - Wen Xi Tang
- Imperial College Healthcare N.H.S. Trust, St Mary’s Hospital, Praed Street, London, United Kingdom
| | - Jackie Man
- Imperial College Healthcare N.H.S. Trust, St Mary’s Hospital, Praed Street, London, United Kingdom
| | - Mingming Yang
- Imperial College Healthcare N.H.S. Trust, St Mary’s Hospital, Praed Street, London, United Kingdom
| | - Erica Zhao
- Imperial College Healthcare N.H.S. Trust, St Mary’s Hospital, Praed Street, London, United Kingdom
| | - Lily Hoque
- Imperial College Healthcare N.H.S. Trust, St Mary’s Hospital, Praed Street, London, United Kingdom
| | - Elif Tuncay
- Imperial College Healthcare N.H.S. Trust, St Mary’s Hospital, Praed Street, London, United Kingdom
| | - Peter Wilding
- Imperial College Healthcare N.H.S. Trust, St Mary’s Hospital, Praed Street, London, United Kingdom
| | - Ian Godsland
- Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London, United Kingdom
| | - Omid Halse
- Imperial College Healthcare N.H.S. Trust, St Mary’s Hospital, Praed Street, London, United Kingdom
| | - Soma Banerjee
- Imperial College Healthcare N.H.S. Trust, St Mary’s Hospital, Praed Street, London, United Kingdom
| | - Nick Oliver
- Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London, United Kingdom
| | - Neil E. Hill
- Imperial College Healthcare N.H.S. Trust, St Mary’s Hospital, Praed Street, London, United Kingdom
| |
Collapse
|
2
|
Meng F, Wang J, Wang L, Zou W. Glucose metabolism impairment in major depressive disorder. Brain Res Bull 2025; 221:111191. [PMID: 39788458 DOI: 10.1016/j.brainresbull.2025.111191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/26/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025]
Abstract
Major depressive disorder (MDD) is a common mental disorder with chronic tendencies that seriously affect regular work, life, and study. However, its exact pathogenesis remains unclear. Patients with MDD experience systemic and localized impairments in glucose metabolism throughout the disease course, disrupting various processes such as glucose uptake, glycoprotein transport, glycolysis, the tricarboxylic acid cycle (TCA), and oxidative phosphorylation (OXPHOS). These impairments may result from mechanisms including insulin resistance, hyperglycemia-induced damage, oxidative stress, astrocyte abnormalities, and mitochondrial dysfunction, leading to insufficient energy supply, altered synaptic plasticity, neuronal cell death, and functional and structural damage to reward networks. These mechanical changes contribute to the pathogenesis of MDD and severely interfere with the prognosis. Herein, we summarized the impairment of glucose metabolism and its pathophysiological mechanisms in patients with MDD. In addition, we briefly discussed potential pharmacological interventions for glucose metabolism to alleviate MDD, including glucagon-like peptide-1 receptor agonists, metformin, topical insulin, liraglutide, and pioglitazone, to encourage the development of new therapeutics.
Collapse
Affiliation(s)
- Fanhao Meng
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, China
| | - Jing Wang
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, China
| | - Long Wang
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, China.
| | - Wei Zou
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, China.
| |
Collapse
|
3
|
Qi L, Geng X, Feng R, Wu S, Fu T, Li N, Ji H, Cheng R, Wu H, Wu D, Huang L, Long Q, Wang X. Association of glycemic variability and prognosis in patients with traumatic brain injury: A retrospective study from the MIMIC-IV database. Diabetes Res Clin Pract 2024; 217:111869. [PMID: 39332533 DOI: 10.1016/j.diabres.2024.111869] [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: 04/10/2024] [Revised: 07/28/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
BACKGROUND Elevated glycemic variability (GV) often occurs in intensive care unit (ICU) patients and is associated with patient prognosis. However, the association between GV and prognosis in ICU patients with traumatic brain injury (TBI) remains unclear. METHOD Clinical data of ICU patients with TBI were obtained from the Medical Information Mart for Intensive Care (MIMIC) -IV database. The coefficient of variation (CV) was utilized to quantify GV, while the Glasgow Coma Scale (GCS) was employed to evaluate the consciousness status of TBI patients. Pearson linear correlation analysis, linear regression, COX regression and restricted cubic spline (RCS) were used to investigate the relationship between CV and consciousness impairment, as well as the risk of in-hospital mortality. RESULT A total of 1641 ICU patients with TBI were included in the study from the MIMIC-IV database. Pearson linear correlation and restricted cubic spline (RCS) analysis results showed a negative linear relationship between CV and the last GCS (P = 0.002) with no evidence of nonlinearity (P for nonlinear = 0.733). Multivariable linear regression suggested a higher CV was associated with a lower discharge GCS [β (95 %CI) = -1.86 (-3.08 ∼ -0.65), P = 0.003]. Furthermore, multivariable COX regression indicated that CV ≥ 0.3 was a risk factor for in-hospital death in TBI patients [HR (95 %CI) = 1.74 (1.15-2.62), P = 0.003], and this result was also consistent across sensitivity and subgroup analyses. CONCLUSION Higher GV is related to poorer consciousness outcomes and increased risk of in-hospital death in ICU patients with TBI. Additional research is needed to understand the logical relationship between GV and TBI progression.
Collapse
Affiliation(s)
- Linrui Qi
- Department of Neurology, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| | - Xin Geng
- Department of Neurosurgery, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| | - Rongliang Feng
- Department of Neurosurgery, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Department of Neurosurgery, the First People's Hospital of Zhaoqing City, Zhaoqing 526060, China.
| | - Shuaishuai Wu
- Department of Neurosurgery, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| | - Tengyue Fu
- Department of Neurosurgery, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| | - Ning Li
- Department of Neurosurgery, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| | - Hongming Ji
- Department of Neurosurgery, Shanxi Provincial People's Hospital, Fifth Hospital of Shanxi Medical University, Shanxi Provincial Key Laboratory of Intelligent, Big Data and Digital Neurosurgery, Shanxi Provincial Key Laboratory of Intelligent Brain Tumor, Taiyuan 030012, China.
| | - Rui Cheng
- Department of Neurosurgery, Shanxi Provincial People's Hospital, Fifth Hospital of Shanxi Medical University, Shanxi Provincial Key Laboratory of Intelligent, Big Data and Digital Neurosurgery, Shanxi Provincial Key Laboratory of Intelligent Brain Tumor, Taiyuan 030012, China.
| | - Hao Wu
- Department of Neurosurgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China.
| | - Dan Wu
- Department of Neurosurgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China.
| | - Lian Huang
- Department of Neurology, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| | - Qingshan Long
- Department of Neurosurgery, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Department of Neurosurgery, Zhongshan Torch Development Zone People's Hospital, Zhongshan 528400, China.
| | - Xiangyu Wang
- Department of Neurosurgery, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| |
Collapse
|
4
|
Cheng L, Yang G, Sun J, Ma J, Fan M. High blood glucose variability may predict poor outcomes in patients with spontaneous cerebellar hemorrhage undergoing surgical operation: a retrospective study. BMC Neurol 2024; 24:244. [PMID: 39009963 PMCID: PMC11247892 DOI: 10.1186/s12883-024-03758-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Elevated blood glucose (BG) variability has been reported as an independent risk factor for poor prognosis in a variety of diseases. This study aimed to investigate the association between BG variability and clinical outcomes in patients with spontaneous cerebellar hemorrhage (SCH) undergoing surgical operation. METHODS This retrospective cohort study of the consecutive patients admitted to the department of Neurosurgery, the Affiliated Hospital of Qingdao University between January 2014 and June 2022 with the diagnosis of SCH underwent surgical intervention. BG analysis was continuously and routinely performed. BG variability was represented by the standard deviation (SD) of the serial measurements within the first 7 days. The general characteristics, imageological information, blood glucose level, and surgical information were reviewed and compared through medical records. RESULTS A total of 115 patients (65 male and 50 female) were enrolled. Out of all 115 patients, the overall clinical outcomes according to the modified Rankin Scale (mRS) were poor (mRS 3-6) in 31 patients (26.96%) and good (mRS 0-2) in 84 patients (73.04%). Twelve of the 115 patients died during hospitalization, and the mortality rate was 10.43%. Multivariate logistic regression analysis showed that SD of BG (odds ratio (OR), 4.717; 95% confidence interval (CI), 1.054-21.115; P = 0.043), GCS (OR, 0.563; 95% CI, 0.330-0.958; P = 0.034), and hematoma volume (OR, 1.395; 95% CI, 1.118-1.748; P = 0.003) were significant predictors. The area under the ROC curve of SD of BG was 0.911 (95% CI, 0.850-0.973; P < 0.001) with a sensitivity and specificity of 90.3% and 83.3%, respectively, and the cut-off value was 1.736. CONCLUSIONS High BG Variability is independently correlated with the 6-month poor outcomes in patients with SCH undergoing surgical operation.
Collapse
Affiliation(s)
- Lei Cheng
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Guifeng Yang
- Department of Radiology, the Third People's Hospital of Qingdao, Qingdao, 266041, China
| | - Jian Sun
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Department of Neurosurgical Intensive Care Unit, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Junwei Ma
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Mingchao Fan
- Department of Neurosurgery, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
- Department of Neurosurgical Intensive Care Unit, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
5
|
Tregub PP, Kulikov VP, Ibrahimli I, Tregub OF, Volodkin AV, Ignatyuk MA, Kostin AA, Atiakshin DA. Molecular Mechanisms of Neuroprotection after the Intermittent Exposures of Hypercapnic Hypoxia. Int J Mol Sci 2024; 25:3665. [PMID: 38612476 PMCID: PMC11011936 DOI: 10.3390/ijms25073665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
The review introduces the stages of formation and experimental confirmation of the hypothesis regarding the mutual potentiation of neuroprotective effects of hypoxia and hypercapnia during their combined influence (hypercapnic hypoxia). The main focus is on the mechanisms and signaling pathways involved in the formation of ischemic tolerance in the brain during intermittent hypercapnic hypoxia. Importantly, the combined effect of hypoxia and hypercapnia exerts a more pronounced neuroprotective effect compared to their separate application. Some signaling systems are associated with the predominance of the hypoxic stimulus (HIF-1α, A1 receptors), while others (NF-κB, antioxidant activity, inhibition of apoptosis, maintenance of selective blood-brain barrier permeability) are mainly modulated by hypercapnia. Most of the molecular and cellular mechanisms involved in the formation of brain tolerance to ischemia are due to the contribution of both excess carbon dioxide and oxygen deficiency (ATP-dependent potassium channels, chaperones, endoplasmic reticulum stress, mitochondrial metabolism reprogramming). Overall, experimental studies indicate the dominance of hypercapnia in the neuroprotective effect of its combined action with hypoxia. Recent clinical studies have demonstrated the effectiveness of hypercapnic-hypoxic training in the treatment of childhood cerebral palsy and diabetic polyneuropathy in children. Combining hypercapnic hypoxia with pharmacological modulators of neuro/cardio/cytoprotection signaling pathways is likely to be promising for translating experimental research into clinical medicine.
Collapse
Affiliation(s)
- Pavel P. Tregub
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
- Brain Science Institute, Research Center of Neurology, 125367 Moscow, Russia
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, RUDN University, 117198 Moscow, Russia; (A.V.V.); (M.A.I.); (A.A.K.); (D.A.A.)
| | - Vladimir P. Kulikov
- Department of Ultrasound and Functional Diagnostics, Altay State Medical University, 656040 Barnaul, Russia;
| | - Irada Ibrahimli
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | | | - Artem V. Volodkin
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, RUDN University, 117198 Moscow, Russia; (A.V.V.); (M.A.I.); (A.A.K.); (D.A.A.)
| | - Michael A. Ignatyuk
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, RUDN University, 117198 Moscow, Russia; (A.V.V.); (M.A.I.); (A.A.K.); (D.A.A.)
| | - Andrey A. Kostin
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, RUDN University, 117198 Moscow, Russia; (A.V.V.); (M.A.I.); (A.A.K.); (D.A.A.)
| | - Dmitrii A. Atiakshin
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, RUDN University, 117198 Moscow, Russia; (A.V.V.); (M.A.I.); (A.A.K.); (D.A.A.)
| |
Collapse
|
6
|
Chi H, Sun Y, Lin P, Zhou J, Zhang J, Yang Y, Qiao Y, Liu D. Glucose Fluctuation Inhibits Nrf2 Signaling Pathway in Hippocampal Tissues and Exacerbates Cognitive Impairment in Streptozotocin-Induced Diabetic Rats. J Diabetes Res 2024; 2024:5584761. [PMID: 38282656 PMCID: PMC10817812 DOI: 10.1155/2024/5584761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 12/25/2023] [Accepted: 01/09/2024] [Indexed: 01/30/2024] Open
Abstract
Background This research investigated whether glucose fluctuation (GF) can exacerbate cognitive impairment in streptozotocin-induced diabetic rats and explored the related mechanism. Methods After 4 weeks of feeding with diets containing high fats plus sugar, the rat model of diabetes mellitus (DM) was established by intraperitoneal injection of streptozotocin (STZ). Then, GF was triggered by means of alternating satiety and starvation for 24 h. The weight, blood glucose level, and water intake of the rats were recorded. The Morris water maze (MWM) test was carried out to appraise the cognitive function at the end of week 12. Moreover, the morphological structure of hippocampal neurons was viewed through HE and Nissl staining, and transmission electron microscopy (TEM) was performed for ultrastructure observation. The protein expression levels of Nrf2, HO-1, NQO-1, Bax, Bcl-2, and Caspase-3 in the hippocampal tissues of rats were measured via Western blotting, and the mRNA expressions of Nrf2, HO-1, and NQO-1 were examined using qRT-PCR. Finally, Western blotting and immunohistochemistry were conducted to detect BDNF levels. Results It was manifested that GF not only aggravated the impairment of spatial memory in rats with STZ-induced type 2 DM but also stimulated the loss, shrinkage, and apoptosis of hippocampal neurons. Regarding the expressions in murine hippocampal tissues, GF depressed Nrf2, HO-1, NQO-1, Bcl-2, and BDNF but boosted Caspase-3 and Bax. Conclusions GF aggravates cognitive impairment by inhibiting the Nrf2 signaling pathway and inducing oxidative stress and apoptosis in the hippocampal tissues.
Collapse
Affiliation(s)
- Haiyan Chi
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Department of Endocrinology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, China
| | - Yujing Sun
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Peng Lin
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Junyu Zhou
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jinbiao Zhang
- Department of Neurology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, China
| | - Yachao Yang
- Department of Endocrinology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, China
| | - Yun Qiao
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Deshan Liu
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
7
|
Schmitt A, Beran M, Geraets A, Iversen MM, Nefs G, Nouwen A, Pouwer F, Huber JW, Schram MT. Associations between HbA1c Reduction and Change in Depressive Symptoms following Glucose-lowering Treatment in Adults: A Systematic Review of Intervention Studies. Curr Diabetes Rev 2024; 20:e020623217607. [PMID: 37278034 DOI: 10.2174/1573399820666230602124223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 04/18/2023] [Accepted: 05/03/2023] [Indexed: 06/07/2023]
Abstract
INTRODUCTION Hyperglycemia constitutes a likely pathway linking diabetes and depressive symptoms; lowering glycemic levels may help reduce diabetes-comorbid depressive symptoms. Since randomized controlled trials can help understand temporal associations, we systematically reviewed the evidence regarding the potential association of hemoglobin HbA1c lowering interventions with depressive symptoms. METHODS PubMed, PsycINFO, CINAHL, and EMBASE databases were searched for randomized controlled trials evaluating HbA1c-lowering interventions and including assessment of depressive symptoms published between 01/2000-09/2020. Study quality was evaluated using the Cochrane Risk of Bias tool. PROSPERO registration: CRD42020215541. RESULTS We retrieved 1,642 studies of which twelve met our inclusion criteria. Nine studies had high and three unclear risks of bias. Baseline depressive symptom scores suggest elevated depressive symptoms in five studies. Baseline HbA1c was <8.0% (<64 mmol/mol) in two, 8.0-9.0% (64-75 mmol/mol) in eight, and ≥10.0% (≥86 mmol/mol) in two studies. Five studies found greater HbA1c reduction in the treatment group; three of these found greater depressive symptom reduction in the treatment group. Of four studies analyzing whether the change in HbA1c was associated with the change in depressive symptoms, none found a significant association. The main limitation of these studies was relatively low levels of depressive symptoms at baseline, limiting the ability to show a lowering in depressive symptoms after HbA1c reduction. CONCLUSIONS We found insufficient available data to estimate the association between HbA1c reduction and depressive symptom change following glucose-lowering treatment. Our findings point to an important gap in the diabetes treatment literature. Future clinical trials testing interventions to improve glycemic outcomes might consider measuring depressive symptoms as an outcome to enable analyses of this association.
Collapse
Affiliation(s)
- Andreas Schmitt
- Research Institute Diabetes Academy Mergentheim (FIDAM), Diabetes Center Mergentheim (DZM), Johann-Hammer- Straße 24, 97980 Bad Mergentheim, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstraße 1, 85764, Oberschleißheim, Germany
| | - Magdalena Beran
- Department of Internal Medicine, School for Cardiovascular Diseases (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, The Netherlands
| | - Anouk Geraets
- Department of Internal Medicine, School for Cardiovascular Diseases (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
- Department of Psychiatry and Neuropsychology, MHeNs School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | - Marjolein M Iversen
- Faculty of Health and Social Sciences, Western Norway University of Applied Sciences, Inndalsveien 28, 5063 Bergen, Norway
| | - Giesje Nefs
- Department of Medical and Clinical Psychology, Center of Research on Psychological and Somatic Disorders (CoRPS), Tilburg University, Warandelaan 2, 5037 AB Tilburg, The Netherlands
- Department of Medical Psychology, Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein 27, 6525 EZ Nijmegen, The Netherlands
- Diabeter, National Treatment and Research Center for Children, Adolescents and Young Adults with Type 1 Diabetes, Blaak 6, 3011 TA Rotterdam, The Netherlands
| | - Arie Nouwen
- Department of Psychology, Middlesex University London, The Burroughs, London NW4 4BT, United Kingdom
- School of Health, Wellbeing and Social Care, The Open University, Walton Hall, Milton Keynes MK7 6AA, United Kingdom
| | - Frans Pouwer
- Department of Psychology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
- Steno Diabetes Center Odense (SDCO), Odense University Hospital, J. B. Winsløws Vej 4, 5000 Odense C, Denmark
| | - Jörg W Huber
- School of Sport and Health Sciences, University of Brighton, Westlain House, Brighton BN1 9PH, United Kingdom
| | - Miranda T Schram
- Department of Internal Medicine, School for Cardiovascular Diseases (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
- Department of Psychiatry and Neuropsychology, MHeNs School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
- Heart and Vascular Centre, Maastricht University Medical Center, Professor Debyelaan 25, 6229HX Maastricht, The Netherlands
| |
Collapse
|
8
|
Schaich CL, Bancks MP, Hayden KM, Ding J, Rapp SR, Bertoni AG, Heckbert SR, Hughes TM, Mongraw-Chaffin M. Visit-to-Visit Glucose Variability, Cognition, and Global Cognitive Decline: The Multi-Ethnic Study of Atherosclerosis. J Clin Endocrinol Metab 2023; 109:e243-e252. [PMID: 37497618 PMCID: PMC10735301 DOI: 10.1210/clinem/dgad444] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/09/2023] [Accepted: 07/26/2023] [Indexed: 07/28/2023]
Abstract
CONTEXT Higher visit-to-visit glucose variability (GV) is associated with dysglycemia and type 2 diabetes (T2D), key risk factors for cognitive decline. OBJECTIVE Evaluate the association of GV with cognitive performance and decline in racially/ethnically diverse older populations with and without T2D. METHODS We calculated the standard deviation of glucose (SDG), average real variability (ARV), and variability independent of the mean (VIM) among 4367 Multi-Ethnic Study of Atherosclerosis participants over 6 clinical examinations. Participants completed a cognitive assessment at the fifth examination, and a subset completed a second assessment 6 years later. We used multivariable linear regression to estimate the association of intraindividual GV with cognitive test scores after adjustments for cardiovascular risk factors and mean glucose level over the study period. RESULTS Two-fold increments in the VIM and SDG were associated with worse Cognitive Abilities Screening Instrument (CASI) performance, while two-fold increments in VIM and ARV were associated with worse Digit Symbol Coding test score. GV measures were not associated with change in CASI performance among 1834 participants with repeat CASI data 6 years later. However, among 229 participants with incident T2D, the SDG and VIM were associated with decline in CASI (-1.7 [95% CI: -3.1, -0.3] and -2.1 [-3.7, -0.5] points, respectively). In contrast, single-timepoint glucose and HbA1c were not associated with CASI decline among participants with or without incident T2D. CONCLUSION Higher visit-to-visit GV over 16 to 18 years is associated with worse cognitive performance in the general population, and with modest global cognitive decline in participants with T2D.
Collapse
Affiliation(s)
- Christopher L Schaich
- Department of Surgery, Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Michael P Bancks
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Kathleen M Hayden
- Department of Social Sciences and Health Policy, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Jingzhong Ding
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Stephen R Rapp
- Department of Psychiatry and Behavioral Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Alain G Bertoni
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Susan R Heckbert
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA 98105, USA
| | - Timothy M Hughes
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Morgana Mongraw-Chaffin
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| |
Collapse
|
9
|
Kwon M, Lee M, Kim EH, Choi DW, Jung E, Kim KY, Jung I, Ha J. Risk of depression and anxiety disorders according to long-term glycemic variability. J Affect Disord 2023; 343:50-58. [PMID: 37734626 DOI: 10.1016/j.jad.2023.09.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/29/2023] [Accepted: 09/17/2023] [Indexed: 09/23/2023]
Abstract
BACKGROUND Poor glycemic control has been linked to psychiatric symptoms. However, studies investigating the relationship between glycemic variability (GV) and depression and anxiety disorders are limited. We investigated the association of GV with depression and anxiety disorders. In addition, the relationship between trends in fasting plasma glucose (FPG) levels and these disorders were explored. METHODS We analyzed the National Health Insurance Service-National Sample Cohort database (2002-2013) with 151,814 participants who had at least three health screenings between 2002 and 2010. Visit-to-visit FPG variability was measured as variability independent of the mean (VIM). Depression and anxiety disorders were diagnosed using ICD-10 codes (F41 for anxiety and F32 or F33 for depression) after index date. We analyzed the association between GV and incidences of these disorders using Kaplan-Meier and Cox proportional hazards methods. Trajectory analysis was conducted to explore the relationship between FPG trends and these disorders. RESULTS During follow-up, 7166 and 14,149 patients were newly diagnosed with depression and anxiety disorders, respectively. The highest quartile group of FPG-VIM had a greater incidence of depression and anxiety than the lowest quartile group, with adjusted hazard ratios of 1.09 (95 % confidence interval [CI]: 1.02-1.17) and 1.08 (95 % CI: 1.03-1.14). Group with persistent hyperglycemia, identified through trajectory clustering of FPG levels, had a 1.43-fold increased risk of depression compared to those with consistently low FPG levels. LIMITATIONS Potential selection bias by including participants with at least three health screenings. CONCLUSIONS High GV and persistent hyperglycemia are associated with increased incidence of depression and anxiety disorders.
Collapse
Affiliation(s)
- Manjae Kwon
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Myeongjee Lee
- Biostatistics Collaboration Unit, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Hwa Kim
- Biostatistics Collaboration Unit, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dong-Woo Choi
- Cancer Big Data Center, National Cancer Control Institute, National Cancer Center, Gyeonggi-do, Republic of Korea
| | - Eunjin Jung
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Keun You Kim
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Psychiatry, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Inkyung Jung
- Division of Biostatistics, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Junghee Ha
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
10
|
Cai W, Li Y, Guo K, Wu X, Chen C, Lin X. Association of glycemic variability with death and severe consciousness disturbance among critically ill patients with cerebrovascular disease: analysis of the MIMIC-IV database. Cardiovasc Diabetol 2023; 22:315. [PMID: 37974159 PMCID: PMC10652479 DOI: 10.1186/s12933-023-02048-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 10/30/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND The association of glycemic variability with severe consciousness disturbance and in-hospital all-cause mortality in critically ill patients with cerebrovascular disease (CVD) remains unclear, This study aimed to investigate the association of glycemic variability with cognitive impairment and in-hospital death. METHOD We extracted all blood glucose measurements of patients diagnosed with CVD from the Medical Information Mart for Intensive Care IV (MIMIC-IV). Glycemic variability was defined as the coefficient of variation (CV), which was determined using the ratio of standard deviation and the mean blood glucose levels. Cox hazard regression models were applied to analyze the link between glycemic variability and outcomes. We also analyzed non-linear relationship between outcome indicators and glycemic variability using restricted cubic spline curves. RESULTS The present study included 2967 patients diagnosed with cerebral infarction and 1842 patients diagnosed with non-traumatic cerebral hemorrhage. Log-transformed CV was significantly related to cognitive impairment and in-hospital mortality, as determined by Cox regression. Increasing log-transformed CV was approximately linearly with the risk of cognitive impairment and in-hospital mortality. CONCLUSION High glycemic variability was found to be an independent risk factor for severe cognitive decline and in-hospital mortality in critically ill patients with CVD. Our study indicated that enhancing stability of glycemic variability may reduced adverse outcomes in patients with severe CVD.
Collapse
Affiliation(s)
- Weimin Cai
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yaling Li
- Department Health Management Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 31000, China
| | - Kun Guo
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiao Wu
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Chao Chen
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, No. 2, Fuxue Lane, Wenzhou, 325000, China.
| | - Xinran Lin
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, No. 2, Fuxue Lane, Wenzhou, 325000, China.
| |
Collapse
|
11
|
Understanding the Combined Effects of High Glucose Induced Hyper-Osmotic Stress and Oxygen Tension in the Progression of Tumourigenesis: From Mechanism to Anti-Cancer Therapeutics. Cells 2023; 12:cells12060825. [PMID: 36980166 PMCID: PMC10047272 DOI: 10.3390/cells12060825] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/03/2023] [Accepted: 02/17/2023] [Indexed: 03/09/2023] Open
Abstract
High glucose (HG), a hallmark of the tumour microenvironment, is also a biomechanical stressor, as it exerts hyper-osmotic stress (HG-HO), but not much is known regarding how tumour cells mechanoadapt to HG-HO. Therefore, this study aimed to delineate the novel molecular mechanisms by which tumour cells mechanoadapt to HG/HG-HO and whether phytochemical-based interference in these mechanisms can generate tumour-cell-selective vulnerability to cell death. Mannitol and L-glucose were used as hyper-osmotic equivalents of high glucose. The results revealed that the tumour cells can efficiently mechanoadapt to HG-HO only in the normoxic microenvironment. Under normoxic HG/HG-HO stress, tumour cells polySUMOylate a higher pool of mitotic driver pH3(Ser10), which translocates to the nucleus and promotes faster cell divisions. On the contrary, acute hypoxia dampens HG/HG-HO-associated excessive proliferation by upregulating sentrin protease SENP7. SENP7 promotes abnormal SUMOylation of pH3(Ser10), thereby restricting its nuclear entry and promoting the M-phase arrest and cell loss. However, the hypoxia-arrested cells that managed to survive showed relapse upon reversal to normoxia as well as upregulation of pro-survival-associated SENP1, and players in tumour growth signalling, autophagy, glycolytic pathways etc. Depletion of SENP1 in both normoxia and hypoxia caused significant loss of tumour cells vs undepleted controls. SENP1 was ascertained to restrict the abnormal SUMOylation of pH3(Ser10) in both normoxia and hypoxia, although not so efficiently in hypoxia, due to the opposing activity of SENP7. Co-treatment with Momordin Ic (MC), a natural SENP1 inhibitor, and Gallic Acid (GA), an inhibitor of identified major pro-tumourigenic signalling (both enriched in Momordica charantia), eliminated surviving tumour cells in normal glucose, HG and HG-HO normoxic and hypoxic microenvironments, suggesting that appropriate and enhanced polySUMOylation of pH3(Ser10) in response to HG/HG-HO stress was attenuated by this treatment along with further dampening of other key tumourigenic signalling, due to which tumour cells could no longer proliferate and grow.
Collapse
|
12
|
Robertson ND, Deacon E, Botha K. A critical review of the relationship between type 1 diabetes mellitus, inhibition, and behavioral management. FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2023; 3:1080415. [PMID: 36992790 PMCID: PMC10012078 DOI: 10.3389/fcdhc.2022.1080415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/30/2022] [Indexed: 02/22/2023]
Abstract
Type 1 diabetes mellitus (T1DM) is a chronic and lifelong condition that requires adequate behavior management in order to meet desired health outcomes. The effects of T1DM on the neurocognitive functioning of affected individuals raise concerns about how the disease may influence executive functioning. Inhibition is a core component of executive functioning, and plays a vital role in self-regulation and the restriction of impulsive behaviors. Inhibition may thus play a vital role in the behavior management of people with T1DM. The aim of this study was to identify current gaps in existing knowledge regarding the relationship between T1DM, inhibition, and behavior management. This study employed a critical review design to analyze and synthesize the current scientific literature. Twelve studies were identified through an appraisal process, and the data extracted were thematically analyzed and integrated. The findings of this study indicate that a possible cycle arises between these three constructs, in which T1DM affects inhibition, inhibition affects behavior management, and poor behavior management affects inhibition. It is recommended that future research should focus more specifically on this relationship.
Collapse
Affiliation(s)
- Neville Dean Robertson
- School of Psychosocial Health, Community Psychosocial Research (COMPRES), North-West University, Potchefstroom, South Africa
| | - Elmari Deacon
- School of Psychosocial Health, Community Psychosocial Research (COMPRES), North-West University, Potchefstroom, South Africa
- OPTENTIA, North-West University, Vanderbijlpark, South Africa
| | - Karel Botha
- School of Psychosocial Health, Community Psychosocial Research (COMPRES), North-West University, Potchefstroom, South Africa
| |
Collapse
|
13
|
Lee DY, Kim J, Park S, Park SY, Yu JH, Seo JA, Kim NH, Yoo HJ, Kim SG, Choi KM, Baik SH, Han K, Kim NH. Fasting Glucose Variability and the Risk of Dementia in Individuals with Diabetes: A Nationwide Cohort Study. Diabetes Metab J 2022; 46:923-935. [PMID: 35609876 PMCID: PMC9723200 DOI: 10.4093/dmj.2021.0346] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/08/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND We investigated whether fasting glucose (FG) variability could predict the risk of dementia. METHODS This cohort study analyzed data from Koreans with diabetes after at least three health examinations by the Korean National Health Insurance Corporation between 2005 and 2010, which included at least one examination between 2009 and 2010. A total of 769,554 individuals were included, excluding those aged <40 years and those with dementia. FG variability was measured using the variability independent of the mean (FG-VIM). The incidence of dementia was defined by the International Classification of Diseases 10th Revision codes and prescription of anti-dementia medication and was subdivided into Alzheimer's disease (AD) and vascular dementia (VD). RESULTS During the 6.9-year follow-up, 54,837, 41,032, and 6,892 cases of all-cause dementia, AD, and VD, respectively, were identified. Cox proportional regression analyses showed that as the FG-VIM quartile increased, the risk of dementia serially increased after adjustment for metabolic factors, income status, and diabetes-related characteristics, including the mean FG. Participants in FG-VIM quartile 4 showed a 18%, 19%, and 17% higher risk for all-cause dementia, AD, and VD, respectively, than those in quartile 1; this particularly included non-obese patients with a longer duration of diabetes, high FG levels, dyslipidemia, and those taking glucose-lowering medications. Conversely, the baseline FG status and dementia showed a U-shaped association. CONCLUSION Increased FG variability over 5 years can predict the risk of dementia in individuals with diabetes in Korea. This finding was more pronounced in patients with less favorable metabolic profiles.
Collapse
Affiliation(s)
- Da Young Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Jaeyoung Kim
- Research Institute for Skin Image, Korea University College of Medicine, Seoul, Korea
- Core Research & Development Center, Korea University Ansan Hospital, Ansan, Korea
| | - Sanghyun Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - So Young Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Ji Hee Yu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Ji A Seo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Nam Hoon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Hye Jin Yoo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Sin Gon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Kyung Mook Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Sei Hyun Baik
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Korea
| | - Nan Hee Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
- BK21 FOUR R&E Center for Learning Health Systems, Korea University, Seoul, Korea
| |
Collapse
|
14
|
Cacciatore M, Grasso EA, Tripodi R, Chiarelli F. Impact of glucose metabolism on the developing brain. Front Endocrinol (Lausanne) 2022; 13:1047545. [PMID: 36619556 PMCID: PMC9816389 DOI: 10.3389/fendo.2022.1047545] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Glucose is the most important substrate for proper brain functioning and development, with an increased glucose consumption in relation to the need of creating new brain structures and connections. Therefore, alterations in glucose homeostasis will inevitably be associated with changes in the development of the Nervous System. Several studies demonstrated how the alteration of glucose homeostasis - both hyper and hypoglycemia- may interfere with the development of brain structures and cognitivity, including deficits in intelligence quotient, anomalies in learning and memory, as well as differences in the executive functions. Importantly, differences in brain structure and functionality were found after a single episode of diabetic ketoacidosis suggesting the importance of glycemic control and stressing the need of screening programs for type 1 diabetes to protect children from this dramatic condition. The exciting progresses of the neuroimaging techniques such as diffusion tensor imaging, has helped to improve the understanding of the effects, outcomes and mechanisms underlying brain changes following dysglycemia, and will lead to more insights on the physio-pathological mechanisms and related neurological consequences about hyper and hypoglycemia.
Collapse
|
15
|
Chen Y, Jiang X, Wang J, Wu Z, Wu Y, Ni Z, Yi H, Lu R. Sensitive Oxidation of Sorbitol-Mediated Fe 2+ by H 2O 2: A Reliable TD-NMR Method for Clinical Blood Glucose Detection. Anal Chem 2021; 93:14153-14160. [PMID: 34637275 DOI: 10.1021/acs.analchem.1c02616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The clinical challenge of high-accuracy blood glucose detection schemes is to overcome the detection error caused by the background interferences in different individuals. H2O2 as the specific product of glucose oxidation can be involved in the Fe2+/Fe3+ conversion and detected by the time-domain nuclear magnetic resonance (TD-NMR) method sensitively. But, in clinical applications, the oxidation of Fe2+ is susceptible to the complex sample substrates. In this work, we sorted out two kinds of possible interference mechanisms of Fe2+ oxidation in the NMR blood glucose detection method and proposed a feasible scheme that uses sorbitol to weaken the adverse effects of interference. We found that sorbitol-mediated Fe2+ can greatly enhance the sensitivity of the T2 value to H2O2. The chain reaction caused by sorbitol can significantly amplify the efficiency of Fe2+ oxidation at the same concentration of H2O2. Thereby, we can achieve the higher dilution multiple of serum samples to reduce the amount of interfering substances involved in the Fe2+/Fe3+ conversion. We justified the accuracy and availability of our method by successfully detecting and confirming the correlation between the T2 decrease and glucose concentration of the serum samples collected from 16 subjects. The sorbitol-Fe2+ glucose detection method with high sensitivity can be further combined with miniature NMR analyzers to satisfy the calibration requirements of glucose monitoring in diabetic patients instead of frequent medical visits.
Collapse
Affiliation(s)
- Yi Chen
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing 211189, People's Republic of China.,School of Mechanical Engineering, Southeast University, Nanjing 211189, People's Republic of China
| | - Xiaowen Jiang
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing 211189, People's Republic of China.,School of Mechanical Engineering, Southeast University, Nanjing 211189, People's Republic of China
| | - Junnan Wang
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing 211189, People's Republic of China.,School of Mechanical Engineering, Southeast University, Nanjing 211189, People's Republic of China
| | - Zhengxiu Wu
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing 211189, People's Republic of China.,School of Mechanical Engineering, Southeast University, Nanjing 211189, People's Republic of China
| | - Yuchen Wu
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing 211189, People's Republic of China.,School of Mechanical Engineering, Southeast University, Nanjing 211189, People's Republic of China
| | - Zhonghua Ni
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing 211189, People's Republic of China.,School of Mechanical Engineering, Southeast University, Nanjing 211189, People's Republic of China
| | - Hong Yi
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing 211189, People's Republic of China.,School of Mechanical Engineering, Southeast University, Nanjing 211189, People's Republic of China
| | - Rongsheng Lu
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, Southeast University, Nanjing 211189, People's Republic of China.,School of Mechanical Engineering, Southeast University, Nanjing 211189, People's Republic of China.,National Key Laboratory of Bioelectronics, Southeast University, Nanjing 211189, People's Republic of China
| |
Collapse
|
16
|
Tang X, Cardoso MA, Yang J, Zhou JB, Simó R. Impact of Intensive Glucose Control on Brain Health: Meta-Analysis of Cumulative Data from 16,584 Patients with Type 2 Diabetes Mellitus. Diabetes Ther 2021; 12:765-779. [PMID: 33548021 PMCID: PMC7947088 DOI: 10.1007/s13300-021-01009-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 01/23/2021] [Indexed: 01/11/2023] Open
Abstract
INTRODUCTION Despite growing evidence that type 2 diabetes is associated with dementia, the question of whether intensive glucose control can prevent or arrest cognitive decline remains unanswered. In the analysis reported here, we explored the effect of intensive glucose control versus standard care on brain health, including structural abnormalities of the brain (atrophy, white matter hyperintensities, lacunar infarction, and cerebral microbleeds), cognitive dysfunction, and risk of dementia. METHODS We searched the PubMed and Embase databases, the Web of Science website, and the Clinicaltrial.gov registry for studies published in English prior to July 2020. Only studies with a randomized controlled trial (RCT) design were considered. We analyzed structural abnormalities of the brain (atrophy, white matter hyperintensities, lacunar infarction, and cerebral microbleeds), cognitive function (cognitive impairment, executive function, memory, attention, and information-processing speed), and dementia (Alzheimer's disease, vascular dementia, and mixed dementia). RESULTS Six studies (5 different RCTs) with 16,584 participants were included in this meta-analysis. One study that compared structural changes between groups receiving intensive versus conventional glucose control measures reported non-significant results. The results of the five studies, comprising four cohorts, indicated a significantly poorer decline in cognitive function in the intensive glucose control group (β - 0.03, 95% confidence interval [CI] - 0.05 to - 0.02) than in the conventional glucose control group. Further subgroup analysis showed a significant difference in the change in cognitive performance in composite cognitive function (β - 0.03, 95% CI - 0.05 to - 0.01) and memory (β - 0.13, 95% CI - 0.25 to - 0.02). One trial evaluated the prevalence of cognitive impairment and dementia between groups receiving intensive and conventional glucose control, respectively, and the differences were insignificant. CONCLUSION This meta-analysis suggests that intensive glucose control in patients with type 2 diabetes can slow down cognitive decline, especially the decline in composite cognition and memory function. However, further studies are necessary to confirm the impact of strict glucose control on structural abnormalities in the brain and the risk of dementia.
Collapse
Affiliation(s)
- Xingyao Tang
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Marly A Cardoso
- Department of Nutrition, School of Public Health, University of Sao Paulo, Sao Paulo, Brazil
| | - Jinkui Yang
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jian-Bo Zhou
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| | - Rafael Simó
- Endocrinology and Nutrition Department, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, Barcelona, Spain
- Diabetes and Metabolism Research Unit, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
17
|
Wang B, Li Y, You C. miR-129-3p Targeting of MCU Protects Against Glucose Fluctuation-Mediated Neuronal Damage via a Mitochondrial-Dependent Intrinsic Apoptotic Pathway. Diabetes Metab Syndr Obes 2021; 14:153-163. [PMID: 33488104 PMCID: PMC7815084 DOI: 10.2147/dmso.s285179] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 12/12/2020] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION Glucose fluctuations have an adverse effect on several diabetes-related complications, especially for the nervous system, but the underlying mechanisms are not clear. MicroRNAs are critical regulators of posttranscription in many physiological processes, such as apoptosis. Our study clarified the neuroprotective effects of miR-129-3p targeting mitochondrial calcium uniporter (MCU) in glucose fluctuation-mediated neuronal damage and the specific mechanisms involved. METHODS The expression of MCU and miR-129-3p was examined by real-time PCR and Western blot in the glucose fluctuation cell model. Dual-luciferase reporter assay was performed to confirm the transcriptional regulation of miR-129-3p by MCU. Fluorescent probe and assay kit assay was used to determine oxidative stress condition. Mitochondrial-dependent intrinsic apoptotic factors were examined by flow cytometry assay, enzyme-linked immunosorbent assay (ELISA), and gene and protein expression assays. RESULTS We found an upregulation of MCU and downregulation of miR-129-3p in glucose fluctuation-treated primary hippocampal neuronal cells, and miR-129-3p directly targeted MCU. miR-129-3p overexpression produced a dramatic reduction in calcium overload, reactive oxygen species (ROS) generation, GSH-to-GSSG ratio, MMP-2 expression in the mitochondrial-dependent intrinsic apoptosis pathway and an increase in MnSOD activity. Increasing MCU expression rescued the effects of miR-129-3p overexpression. miR-129-3p downregulation produced a significant increase in calcium overload, reactive oxygen species (ROS) generation, MMP-2 expression, cytochrome c release and cell apoptosis, and antioxidant N-acetyl cysteine (NAC) rescued the effects of miR-129-3p downregulation. CONCLUSION Therefore, miR-129-3p suppressed glucose fluctuation-mediated neuronal damage by targeting MCU via a mitochondrial-dependent intrinsic apoptotic pathway. The miR-129-3p/MCU axis may be a promising therapeutic target for glucose fluctuation-mediated neuronal damage.
Collapse
Affiliation(s)
- Bo Wang
- Department of Neurosurgery, West China School of Medicine/West China Hospital of Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
- Department of Neurosurgery, Kunming Medical University First Affiliated Hospital, Kunming, Yunnan650032, People’s Republic of China
| | - Yang Li
- Intensive Care Unit, West China School of Medicine/West China Hospital of Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
| | - Chao You
- Department of Neurosurgery, West China School of Medicine/West China Hospital of Sichuan University, Chengdu, Sichuan610041, People’s Republic of China
- Correspondence: Chao You Department of Neurosurgery, West China School of Medicine/West China Hospital of Sichuan University, Chengdu, Sichuan610041, People’s Republic of ChinaTel +86 28-85422026 Email
| |
Collapse
|
18
|
Watt C, Sanchez-Rangel E, Hwang JJ. Glycemic Variability and CNS Inflammation: Reviewing the Connection. Nutrients 2020; 12:nu12123906. [PMID: 33371247 PMCID: PMC7766608 DOI: 10.3390/nu12123906] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/16/2022] Open
Abstract
Glucose is the primary energy source for the brain, and exposure to both high and low levels of glucose has been associated with numerous adverse central nervous system (CNS) outcomes. While a large body of work has highlighted the impact of hyperglycemia on peripheral and central measures of oxidative stress, cognitive deficits, and vascular complications in Type 1 and Type 2 diabetes, there is growing evidence that glycemic variability significantly drives increased oxidative stress, leading to neuroinflammation and cognitive dysfunction. In this review, the latest data on the impact of glycemic variability on brain function and neuroinflammation will be presented. Because high levels of oxidative stress have been linked to dysfunction of the blood-brain barrier (BBB), special emphasis will be placed on studies investigating the impact of glycemic variability on endothelial and vascular inflammation. The latest clinical and preclinical/in vitro data will be reviewed, and clinical/therapeutic implications will be discussed.
Collapse
|
19
|
Resveratrol Prevents GLUT3 Up-Regulation Induced by Middle Cerebral Artery Occlusion. Brain Sci 2020; 10:brainsci10090651. [PMID: 32962200 PMCID: PMC7563146 DOI: 10.3390/brainsci10090651] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/07/2020] [Accepted: 09/18/2020] [Indexed: 12/21/2022] Open
Abstract
Glucose transporter (GLUT)3 up-regulation is an adaptive response activated to prevent cellular damage when brain metabolic energy is reduced. Resveratrol is a natural polyphenol with anti-oxidant and anti-inflammatory features that protects neurons against damage induced in cerebral ischemia. Since transcription factors sensitive to oxidative stress and inflammation modulate GLUT3 expression, the purpose of this work was to assess the effect of resveratrol on GLUT3 expression levels after ischemia. Male Wistar rats were subjected to 2 h of middle cerebral artery occlusion (MCAO) followed by different times of reperfusion. Resveratrol (1.9 mg/kg; i. p.) was administered at the onset of the restoration of the blood flow. Quantitative-PCR and Western blot showed that MCAO provoked a substantial increase in GLUT3 expression in the ipsilateral side to the lesion of the cerebral cortex. Immunofluorescence assays indicated that GLUT3 levels were upregulated in astrocytes. Additionally, an important increase in GLUT3 occurred in other cellular types (e.g., damaged neurons, microglia, or infiltrated macrophages). Immunodetection of the microtubule-associated protein 2 (MAP2) showed that MCAO induced severe damage to the neuronal population. However, the administration of resveratrol at the time of reperfusion resulted in injury reduction. Resveratrol also prevented the MCAO-induced increase of GLUT3 expression. In conclusion, resveratrol protects neurons from damage induced by ischemia and prevents GLUT3 upregulation in the damaged brain that might depend on AMPK activation.
Collapse
|
20
|
Xiong Z, Li J, Zhong X, Zhang S, Sun X, Zhou H, Chen Y, Zhuang X, Liao X. Visit-to-Visit Fasting Glucose Variability in Young Adulthood and Hippocampal Integrity and Volume at Midlife. Diabetes Care 2019; 42:2334-2337. [PMID: 31548243 PMCID: PMC7364671 DOI: 10.2337/dc19-0834] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 08/30/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To determine whether visit-to-visit fasting glucose (VVFG) variability in young adulthood is associated with midlife hippocampal integrity and volume. RESEARCH DESIGN AND METHODS Multivariable-adjusted linear regression models were used to estimate the association between VVFG variability and brain MRI variables in 543 CARDIA study participants. VVFG variability was defined by the SD of FG (SDFG), the coefficient of variation of the mean FG (CVFG), and the average real variability (ARVFG) over 25 years of follow-up. Hippocampal integrity fractional anisotropy (FA) and tissue volume standardized to intracranial volume were measured by 3T MRI at year 25. RESULTS After multivariable adjustment, higher FG variability (1-SD increase) was associated with lower hippocampal FA (SDFG -0.015 [95% CI -0.026, -0.004]; CVFG -0.009 [95% CI -0.018, -0.001]; ARVFG -0.011 [95% CI -0.019, -0.002]) and lower hippocampal volume (SDFG -0.012 [95% CI -0.023, -0.001]). CONCLUSIONS Higher VVFG variability in young adulthood is associated with lower midlife hippocampal integrity and volume, suggesting its value in predicting risk for hippocampal structural damage.
Collapse
Affiliation(s)
- Zhenyu Xiong
- The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory on Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Jiaying Li
- Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Xiangbin Zhong
- The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory on Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Shaozhao Zhang
- The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory on Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Xiuting Sun
- The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory on Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Huimin Zhou
- The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory on Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Yunzhi Chen
- The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaodong Zhuang
- The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory on Assisted Circulation, Sun Yat-sen University, Guangzhou, China
- Center for Information Technology & Statistics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xinxue Liao
- The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory on Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
21
|
Thalange N, Biester T, Danne T. Clinical Use of Degludec in Children and Adolescents with T1D: A Narrative Review with Fictionalized Case Reports. Diabetes Ther 2019; 10:1219-1237. [PMID: 31187420 PMCID: PMC6612349 DOI: 10.1007/s13300-019-0641-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Indexed: 12/11/2022] Open
Abstract
The use of insulin in children and adolescents with type 1 diabetes (T1D) is a challenge because of the heterogeneity of these patients and their lifestyles, with consequent unpredictability in blood glucose levels. A new ultra-long-acting basal insulin, insulin degludec (degludec), has the potential to mitigate some of these challenges, notably variability in the glucose-lowering action of the basal insulin component of an insulin regimen, and consequent risks of hypo- and hyperglycemia. However, the protracted half-life and steady state pharmacokinetics of degludec potentially bring some new challenges. In particular, the adjustment of therapy in response to commonly encountered clinical situations might require a different approach when degludec is used in place of other currently used basal insulins in this challenging patient population. The purpose of this article is to guide clinicians through a series of case histories in the use of this insulin. These include, but are not limited to, how to initiate, titrate, switch from other basal insulin or pump therapy; how to alleviate difficulties arising as a result of unpredictable lifestyle/habits; and how to maintain treatment following diabetic ketoacidosis. The guidance presented in this review illustrates that degludec is a good option for a diverse range of children and adolescents with T1D, providing much needed flexibility in the treatment of this challenging patient population.Funding Novo Nordisk.
Collapse
Affiliation(s)
- Nandu Thalange
- Al Jalila Children's Specialty Hospital, Al Jaddaf, Dubai, United Arab Emirates.
| | - Torben Biester
- Diabetes Centre for Children and Adolescents, AUF DER BULT, Hannover, Germany
| | - Thomas Danne
- Diabetes Centre for Children and Adolescents, AUF DER BULT, Hannover, Germany
| |
Collapse
|
22
|
Bancks MP, Carnethon MR, Jacobs DR, Launer LJ, Reis JP, Schreiner PJ, Shah RV, Sidney S, Yaffe K, Yano Y, Allen NB. Fasting Glucose Variability in Young Adulthood and Cognitive Function in Middle Age: The Coronary Artery Risk Development in Young Adults (CARDIA) Study. Diabetes Care 2018; 41:2579-2585. [PMID: 30305344 PMCID: PMC6245206 DOI: 10.2337/dc18-1287] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/12/2018] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To determine whether intraindividual variability in fasting glucose (FG) below the threshold of diabetes is associated with cognitive function in middle adulthood beyond increasing FG. RESEARCH DESIGN AND METHODS We studied 3,307 CARDIA (Coronary Artery Risk Development in Young Adults) Study participants (age range 18-30 years and enrolled in 1985-1986) at baseline and calculated two measures of long-term glucose variability: the coefficient of variation about the mean FG (CV-FG) and the absolute difference between successive FG measurements (average real variability [ARV-FG]) before the onset of diabetes over 25 and 30 years of follow-up. Cognitive function was assessed at years 25 (2010-2011) and 30 (2015-2016) with the Digit Symbol Substitution Test (DSST), Rey-Auditory Verbal Learning Test (RAVLT), Stroop Test, Montreal Cognitive Assessment, and category and letter fluency tests. We estimated the association between glucose variability and cognitive function test score with adjustment for clinical and behavioral risk factors, mean FG level, change in FG level, and diabetes development, medication use, and duration. RESULTS After multivariable adjustment, 1-SD increment of CV-FG was associated with worse cognitive scores at year 25: DSST, standardized regression coefficient -0.95 (95% CI -1.54, -0.36); RAVLT, -0.14 (95% CI -0.27, -0.02); and Stroop Test, 0.49 (95% CI 0.04, 0.94). Findings were similar between CV-FG with each cognitive test score at year 30 and when we used an alternative measure of variability (ARV-FG) that captures variability in successive FG values. CONCLUSIONS Higher intraindividual FG variability during young adulthood below the threshold of diabetes was associated with worse processing speed, memory, and language fluency in midlife independent of FG levels.
Collapse
Affiliation(s)
- Michael P Bancks
- Wake Forest University Health Sciences, Winston-Salem, NC
- Northwestern University, Chicago, IL
| | | | | | | | - Jared P Reis
- National Heart, Lung, and Blood Institute, Bethesda, MD
| | | | | | | | - Kristine Yaffe
- University of California, San Francisco, San Francisco, CA
| | | | | |
Collapse
|
23
|
Kim YK, Nam KI, Song J. The Glymphatic System in Diabetes-Induced Dementia. Front Neurol 2018; 9:867. [PMID: 30429819 PMCID: PMC6220044 DOI: 10.3389/fneur.2018.00867] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 09/26/2018] [Indexed: 12/14/2022] Open
Abstract
The glymphatic system has emerged as an important player in central nervous system (CNS) diseases, by regulating the vasculature impairment, effectively controlling the clearance of toxic peptides, modulating activity of astrocytes, and being involved in the circulation of neurotransmitters in the brain. Recently, several studies have indicated decreased activity of the glymphatic pathway under diabetes conditions such as in insulin resistance and hyperglycemia. Furthermore, diabetes leads to the disruption of the blood-brain barrier and decrease of apolipoprotein E (APOE) expression and the secretion of norepinephrine in the brain, involving the impairment of the glymphatic pathway and ultimately resulting in cognitive decline. Considering the increased prevalence of diabetes-induced dementia worldwide, the relationship between the glymphatic pathway and diabetes-induced dementia should be investigated and the mechanisms underlying their relationship should be discussed to promote the development of an effective therapeutic approach in the near future. Here, we have reviewed recent evidence for the relationship between glymphatic pathway dysfunction and diabetes. We highlight that the enhancement of the glymphatic system function during sleep may be beneficial to the attenuation of neuropathology in diabetes-induced dementia. Moreover, we suggest that improving glymphatic system activity may be a potential therapeutic strategy for the prevention of diabetes-induced dementia.
Collapse
Affiliation(s)
- Young-Kook Kim
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, South Korea.,Department of Biomedical Sciences, Center for Creative Biomedical Scientists, Chonnam National University, Gwangju, South Korea
| | - Kwang Il Nam
- Department of Anatomy, Chonnam National University Medical School, Gwangju, South Korea
| | - Juhyun Song
- Department of Biomedical Sciences, Center for Creative Biomedical Scientists, Chonnam National University, Gwangju, South Korea.,Department of Anatomy, Chonnam National University Medical School, Gwangju, South Korea
| |
Collapse
|
24
|
Kazkayasi I, Burul-Bozkurt N, Ismail MAM, Merino-Serrais P, Pekiner C, Cedazo-Minguez A, Uma S. Insulin deprivation decreases insulin degrading enzyme levels in primary cultured cortical neurons and in the cerebral cortex of rats with streptozotocin-induced diabetes. Pharmacol Rep 2018; 70:677-683. [PMID: 29940507 DOI: 10.1016/j.pharep.2018.01.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 12/21/2017] [Accepted: 01/30/2018] [Indexed: 02/01/2023]
Abstract
BACKGROUND Many studies have indicated a relationship between diabetes and Alzheimer's disease (AD). However, the molecular mechanism underlying this association has not been clarified. Among several factors, insulin degrading enzyme (IDE), which plays roles in the degradation of both insulin and amyloid β (Aβ), has gained interest as a potential target in efforts to solve this puzzle. This study sought to examine the effects of varying insulin and/or glucose concentrations on IDE expression. METHODS Experiments were performed on primary cultured rat neurons and cortices of rats with streptozotocin (STZ)-induced diabetes. IDE protein and mRNA expression levels were measured by western blot and RT-PCR, respectively. RESULTS In primary cultured cortical neurons, removal of insulin for 5days reduced the expression of IDE. A five-day treatment with a high concentration of glucose in insulin-free media reduced IDE levels, while a high concentration of glucose in the presence of insulin had no effect. In groups treated with glucose or insulin intermittently, the reduction in IDE levels was observed only in neurons exposed to high glucose together with no insulin for 5days. Shorter incubation periods (48h), either continuously or intermittently, did not affect IDE levels. IDE expression in the cortex of rats with STZ-induced diabetes was found to be decreased. CONCLUSION Our data suggest that insulin deprivation, rather than high glucose, is a significant determinant of IDE regulation. As evidence indicates potential roles for IDE in diabetes and AD, understanding the mechanisms regulating IDE expression may be important in developing new treatment strategies.
Collapse
Affiliation(s)
- Inci Kazkayasi
- Hacettepe University, Faculty of Pharmacy, Department of Pharmacology, Ankara, Turkey.
| | - Nihan Burul-Bozkurt
- Hacettepe University, Faculty of Pharmacy, Department of Pharmacology, Ankara, Turkey
| | - Muhammad-Al-Mustafa Ismail
- Karolinska Institute, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division for Neurogeriatrics, Huddinge, Sweden
| | - Paula Merino-Serrais
- Karolinska Institute, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division for Neurogeriatrics, Huddinge, Sweden
| | - Can Pekiner
- Hacettepe University, Faculty of Pharmacy, Department of Pharmacology, Ankara, Turkey
| | - Angel Cedazo-Minguez
- Karolinska Institute, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division for Neurogeriatrics, Huddinge, Sweden
| | - Serdar Uma
- Hacettepe University, Faculty of Pharmacy, Department of Pharmacology, Ankara, Turkey
| |
Collapse
|
25
|
Nilsson M, Gjedde A, Brock B, Gejl M, Rungby J. The effects of incretin hormones on cerebral glucose metabolism in health and disease. Neuropharmacology 2017; 136:243-250. [PMID: 29274367 DOI: 10.1016/j.neuropharm.2017.12.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 12/14/2017] [Accepted: 12/18/2017] [Indexed: 12/25/2022]
Abstract
Incretin hormones, notably glucagon-like peptide-1 (GLP-1), are gluco-regulatory hormones with pleiotropic effects also in the central nervous system. Apart from a local production of GLP-1, systemic administration of the hormone has been shown to influence a number of cerebral pathologies, including neuroinflammation. Given the brains massive dependence on glucose as its major fuel, we here review the mechanistics of cerebral glucose transport and metabolism, focusing on the deleterious effects of both hypo- and hyperglycaemia. GLP-1, when administered as long-acting analogues or intravenously, appears to decrease transport of glucose in normoglycaemic conditions, without affecting the total cerebral glucose content. During hypoglycaemia this effect seems abated, whereas during hyperglycaemia GLP-1 regulates cerebral glucose metabolism towards stable levels resembling normoglycaemia. In Alzheimer's disease, a 6-month intervention with GLP-1 maintained cerebral glucose levels at baseline levels, contrasting the decline otherwise seen in Alzheimer's. Kinetic studies suggest blood-brain barrier (BBB) glucose transport as the key player in GLP-1 mediated effects on cerebral glucose metabolism. This article is part of the Special Issue entitled 'Metabolic Impairment as Risk Factors for Neurodegenerative Disorders.'
Collapse
Affiliation(s)
- Malin Nilsson
- Department of Endocrinology, Bispebjerg University Hospital, Copenhagen, Denmark
| | - Albert Gjedde
- Department of Neuroscience, Panum Institute, University of Copenhagen, Copenhagen, Denmark; Departments of Clinical Research, and Department of Nuclear Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Michael Gejl
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Jørgen Rungby
- Department of Endocrinology, Bispebjerg University Hospital, Copenhagen, Denmark.
| |
Collapse
|
26
|
Piechowiak K, Dżygało K, Szypowska A. The additional dose of insulin for high-protein mixed meal provides better glycemic control in children with type 1 diabetes on insulin pumps: randomized cross-over study. Pediatr Diabetes 2017; 18:861-868. [PMID: 28117542 DOI: 10.1111/pedi.12500] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Revised: 11/26/2016] [Accepted: 12/16/2016] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Delivery of insulin for high-protein low-fat meals with carbohydrates on the basis of carbohydrates leads to higher late postprandial glycemia. Studies with mixed meals demonstrated lower blood glucose level after dual wave bolus. The objective of our study was to assess the impact of additional dose of insulin in dual wave bolus for high-protein mixed meal on the postprandial glycemia. MATERIALS AND METHODS We performed a randomized, double-blind, two-way cross-over study, including 58 children with type 1 diabetes, aged 14.7 ± 2.2 years. Participants were randomly assigned into two treatment orders: NORMAL-DUAL or DUAL-NORMAL BOLUS. They consumed standardized high-protein, low-fat meals with carbohydrates. The primary outcome was postprandial glycemia (PPG) based on capillary blood glucose measurements (CBGM). The secondary outcomes were the frequency of hypoglycemia, area under glucose curve, mean amplitude of glycemic excursion (MAGE) and glycemic rise. RESULTS PPG assessed at 180 min was significantly lower when dual wave bolus was delivered (NORMAL 162 mg/dL [9 mmol/L] vs DUAL 130.0 mg/dL [7.22 mmol/L]; P = .004). There were no differences in CBGM between both groups at 60 and 120 min. We found differences between the groups in MAGE at 120 min (NORMAL 82.86 mg/dL [4.6 mmol/L] versus DUAL 54.76 mg/dL [3.04 mmol/L]; P = .0008). We observed no differences in the number of hypoglycemic episodes in both groups. CONCLUSION Applying an additional dose of insulin in dual wave bolus for high-protein mixed meal improved PPG. We observed no statistically significant increase in the number of hypoglycemic episodes associated with this intervention.
Collapse
Affiliation(s)
| | - Katarzyna Dżygało
- Department of Paediatrics, Medical University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
27
|
Engin AB, Engin ED, Karakus R, Aral A, Gulbahar O, Engin A. N-Methyl-D aspartate receptor-mediated effect on glucose transporter-3 levels of high glucose exposed-SH-SY5Y dopaminergic neurons. Food Chem Toxicol 2017; 109:465-471. [PMID: 28951307 DOI: 10.1016/j.fct.2017.09.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/16/2017] [Accepted: 09/20/2017] [Indexed: 11/29/2022]
Abstract
High glucose and insulin lead to neuronal insulin resistance. Glucose transport into the neurons is achieved by regulatory induction of surface glucose transporter-3 (GLUT3) instead of the insulin. N-methyl-D aspartate (NMDA) receptor activity increases GLUT3 expression. This study explored whether an endogenous NMDA receptor antagonist, kynurenic acid (KynA) affects the neuronal cell viability at high glucose concentrations. SH-SY5Y neuroblastoma cells were exposed to 150-250 mg/dL glucose and 40 μU/mL insulin. In KynA and N-nitro-l-arginine methyl ester (L-NAME) supplemented cultures, oxidative stress, mitochondrial metabolic activity (MTT), nitric oxide as nitrite+nitrate (NOx) and GLUT3 were determined at the end of 24 and 48-h incubation periods. Viable cells were counted by trypan blue dye. High glucose-exposed SH-SY5Y cells showed two-times more GLUT3 expression at second 24-h period. While GLUT3-stimulated glucose transport and oxidative stress was increased, total mitochondrial metabolic activity was significantly reduced. Insulin supplementation to high glucose decreased NOx synthesis and GLUT3 levels, in contrast oxidative stress increased three-fold. KynA significantly reduced oxidative stress, and increased MTT by regulating NOx production and GLUT3 expression. KynA is a noteworthy compound, as an endogenous, specific NMDA receptor antagonist; it significantly reduces oxidative stress, while increasing cell viability at high glucose and insulin concentrations.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Gazi University, Faculty of Pharmacy, Department of Toxicology, Hipodrom, Ankara, Turkey.
| | - Evren Doruk Engin
- Ankara University, Biotechnology Institute, Tandogan, Ankara, Turkey
| | - Resul Karakus
- Gazi University, Faculty of Medicine, Department of Immunology, Besevler, Ankara, Turkey
| | - Arzu Aral
- Gazi University, Faculty of Medicine, Department of Immunology, Besevler, Ankara, Turkey
| | - Ozlem Gulbahar
- Gazi University, Faculty of Medicine, Department of Biochemistry, Besevler, Ankara, Turkey
| | - Atilla Engin
- Gazi University, Faculty of Medicine, Department of General Surgery, Besevler, Ankara, Turkey
| |
Collapse
|
28
|
Uncoupling Protein 2 Inhibition Exacerbates Glucose Fluctuation-Mediated Neuronal Effects. Neurotox Res 2017; 33:388-401. [PMID: 28875237 DOI: 10.1007/s12640-017-9805-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/04/2017] [Accepted: 08/23/2017] [Indexed: 01/10/2023]
Abstract
Though glucose fluctuations have been considered as an adverse factor for the development of several diabetes-related complications, their impact in the central nervous system is still not fully elucidated. This study was conducted to evaluate the responses of neuronal cells to different glycemic exposures alongside to elucidate the role of uncoupling protein 2 (UCP2) in regulating such responses. To achieve our goals, primary cortical neurons were submitted to constant high (HG)/low (LG) or glucose level variations (GVs), and the pharmacological inhibition of UCP2 activity was performed using genipin. Results obtained show that GV decreased neuronal cells' viability, mitochondrial membrane potential, and manganese superoxide dismutase activity and increased reactive oxygen species (ROS) production. GV also caused an increase in the glutathione/glutathione disulfide ratio and in the protein expression levels of nuclear factor E2-related factor 2 (NRF2), UCP2, NADH-ubiquinone oxidoreductase chain 1 (ND1), and mitochondrially encoded cytochrome c oxidase I (MTCO1), both mitochondrial DNA encoded subunits of the electron transport chain. Contrariwise, genipin abrogated all those compensations and increased the levels of caspase 3-like activity, potentiated mitochondrial ROS levels, and the loss of neuronal synaptic integrity, decreased the protein expression levels of NRF1, and increased the protein expression levels of UCP5. Further, in the control and LG conditions, genipin increased mitochondrial ROS and the protein expression levels of UCP4, postsynaptic density protein 95 (PSD95), ND1, and MTCO1. Overall, these observations suggest that UCP2 is in the core of neuronal cell protection and/or adaptation against GV-mediated effects and that other isoforms of neuronal UCPs can be upregulated to compensate the inhibition of UCP2 activity.
Collapse
|
29
|
Rawlings AM, Sharrett AR, Mosley TH, Ballew SH, Deal JA, Selvin E. Glucose Peaks and the Risk of Dementia and 20-Year Cognitive Decline. Diabetes Care 2017; 40:879-886. [PMID: 28500217 PMCID: PMC5481977 DOI: 10.2337/dc16-2203] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 03/30/2017] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Hemoglobin A1c (HbA1c), a measure of average blood glucose level, is associated with the risk of dementia and cognitive impairment. However, the role of glycemic variability or glucose excursions in this association is unclear. We examined the association of glucose peaks in midlife, as determined by the measurement of 1,5-anhydroglucitol (1,5-AG) level, with the risk of dementia and 20-year cognitive decline. RESEARCH DESIGN AND METHODS Nearly 13,000 participants from the Atherosclerosis Risk in Communities (ARIC) study were examined. Dementia was ascertained from surveillance, neuropsychological testing, telephone calls with participants or their proxies, or death certificate dementia codes. Cognitive function was assessed using three neuropsychological tests at three visits over 20 years and was summarized as z scores. We used Cox and linear mixed-effects models. 1,5-AG level was dichotomized at 10 μg/mL and examined within clinical categories of HbA1c. RESULTS Over a median time of 21 years, dementia developed in 1,105 participants. Among persons with diabetes, each 5 μg/mL decrease in 1,5-AG increased the estimated risk of dementia by 16% (hazard ratio 1.16, P = 0.032). For cognitive decline among participants with diabetes and HbA1c <7% (53 mmol/mol), those with glucose peaks had a 0.19 greater z score decline over 20 years (P = 0.162) compared with those without peaks. Among participants with diabetes and HbA1c ≥7% (53 mmol/mol), those with glucose peaks had a 0.38 greater z score decline compared with persons without glucose peaks (P < 0.001). We found no significant associations in persons without diabetes. CONCLUSIONS Among participants with diabetes, glucose peaks are a risk factor for cognitive decline and dementia. Targeting glucose peaks, in addition to average glycemia, may be an important avenue for prevention.
Collapse
Affiliation(s)
- Andreea M Rawlings
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - A Richey Sharrett
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Thomas H Mosley
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS
| | - Shoshana H Ballew
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Jennifer A Deal
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
30
|
Diabetes-induced abnormalities of mitochondrial function in rat brain cortex: the effect of n-3 fatty acid diet. Mol Cell Biochem 2017; 435:109-131. [DOI: 10.1007/s11010-017-3061-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 05/04/2017] [Indexed: 01/07/2023]
|
31
|
ROS Production and ERK Activity Are Involved in the Effects of d-β-Hydroxybutyrate and Metformin in a Glucose Deficient Condition. Int J Mol Sci 2017; 18:ijms18030674. [PMID: 28335557 PMCID: PMC5372684 DOI: 10.3390/ijms18030674] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 03/11/2017] [Accepted: 03/16/2017] [Indexed: 12/12/2022] Open
Abstract
Hypoglycemia, a complication of insulin or sulfonylurea therapy in diabetic patients, leads to brain damage. Furthermore, glucose replenishment following hypoglycemic coma induces neuronal cell death. In this study, we investigated the molecular mechanism underlying glucose deficiency-induced cytotoxicity and the protective effect of d-β-hydroxybutyrate (D-BHB) using SH-SY5Y cells. The cytotoxic mechanism of metformin under glucose deficiency was also examined. Cell viability under 1 mM glucose (glucose deficiency) was significantly decreased which was accompanied by increased production of reactive oxygen species (ROS) and decreased phosphorylation of extracellular signal-regulated kinase (ERK) and glycogen synthase 3 (GSK3β). ROS inhibitor reversed the glucose deficiency-induced cytotoxicity and restored the reduced phosphorylation of ERK and GSK3β. While metformin did not alter cell viability in normal glucose media, it further increased cell death and ROS production under glucose deficiency. However, D-BHB reversed cytotoxicity, ROS production, and the decrease in phosphorylation of ERK and GSK3β induced by the glucose deficiency. ERK inhibitor reversed the D-BHB-induced increase in cell viability under glucose deficiency, whereas GSK3β inhibitor did not restore glucose deficiency-induced cytotoxicity. Finally, the protective effect of D-BHB against glucose deficiency was confirmed in primary neuronal cells. We demonstrate that glucose deficiency-induced cytotoxicity is mediated by ERK inhibition through ROS production, which is attenuated by D-BHB and intensified by metformin.
Collapse
|
32
|
Kazkayasi I, Ismail MAM, Parrado-Fernandez C, Björkhem I, Pekiner C, Uma S, Cedazo-Minguez A, Burul-Bozkurt N. Lack of insulin results in reduced seladin-1 expression in primary cultured neurons and in cerebral cortex of STZ-induced diabetic rats. Neurosci Lett 2016; 633:174-181. [PMID: 27639960 DOI: 10.1016/j.neulet.2016.09.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/17/2016] [Accepted: 09/13/2016] [Indexed: 02/01/2023]
Abstract
Several studies demonstrated that Diabetes mellitus (DM) enhances the risk for Alzheimer's disease (AD). Although hyperglycemia and perturbed function of insulin signaling have been proposed to contribute to AD pathogenesis, the molecular mechanisms behind this association is not clear yet. Seladin-1 is an enzyme catalyzing the last step in cholesterol biosynthesis converting desmosterol to cholesterol. The neuroprotective function of seladin-1 has gained interest in AD research recently. Seladin-1 has anti-apoptotic properties and regulates the expression of β-secretase (BACE-1). Here we measured seladin-1 mRNA and protein expressions in rat primary cultured neurons under diabetic conditions and also in the brains of rats with streptozotocine (STZ)-induced diabetes. We show that constant lack of insulin for 5days decreased seladin-1 levels in cultured rat primary neurons. Similarly, a decrease in seladin-1 was found in the brains of rats with STZ-induced diabetes. However, if the lack of insulin and/or high glucose treatment was intermittent, neuronal seladin-1 levels were not affected in vitro. On the other hand, treatment of neurons with metformin resulted in a significant increase in seladin-1. Constant lack of insulin for 5days, as well as high glucose treatment, increased the neuronal expression of BACE-1 in vitro, but not in the in vivo model. Our study defines insulin as a regulator of seladin-1 expression for the first time. The relevance of these findings for the association of DM with AD is discussed.
Collapse
Affiliation(s)
- Inci Kazkayasi
- Hacettepe University, Faculty of Pharmacy, Department of Pharmacology, 06100, Sihhiye, Ankara, Turkey.
| | - Muhammad-Al-Mustafa Ismail
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division for Neurogeriatrics, Karolinska Institute, 14157, Huddinge, Sweden
| | - Cristina Parrado-Fernandez
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division for Neurogeriatrics, Karolinska Institute, 14157, Huddinge, Sweden
| | - Ingemar Björkhem
- Department of Laboratory Medicine, Division of Clinical Chemistry, Karolinska University Hospital, Huddinge, Sweden
| | - Can Pekiner
- Hacettepe University, Faculty of Pharmacy, Department of Pharmacology, 06100, Sihhiye, Ankara, Turkey
| | - Serdar Uma
- Hacettepe University, Faculty of Pharmacy, Department of Pharmacology, 06100, Sihhiye, Ankara, Turkey
| | - Angel Cedazo-Minguez
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division for Neurogeriatrics, Karolinska Institute, 14157, Huddinge, Sweden
| | - Nihan Burul-Bozkurt
- Hacettepe University, Faculty of Pharmacy, Department of Pharmacology, 06100, Sihhiye, Ankara, Turkey
| |
Collapse
|
33
|
McNeilly AD, Gallagher JR, Dinkova-Kostova AT, Hayes JD, Sharkey J, Ashford MLJ, McCrimmon RJ. Nrf2-Mediated Neuroprotection Against Recurrent Hypoglycemia Is Insufficient to Prevent Cognitive Impairment in a Rodent Model of Type 1 Diabetes. Diabetes 2016; 65:3151-60. [PMID: 27411381 DOI: 10.2337/db15-1653] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 07/07/2016] [Indexed: 11/13/2022]
Abstract
It remains uncertain whether recurrent nonsevere hypoglycemia (Hypo) results in long-term cognitive impairment in type 1 diabetes (T1D). This study tested the hypothesis that specifically in the T1D state, Hypo leads to cognitive impairment via a pathological response to oxidative stress. Wild-type (Control) and nuclear factor-erythroid 2 p45-related factor 2 (Nrf2) null mice were studied. Eight groups of mice (Control and Nrf2(-/-) ± T1D and ± Hypo) were subject to recurrent, twice-weekly, insulin or saline injections over 4 weeks, after which cognitive function was assessed and brain tissue analyzed. Recurrent moderate hypoglycemia in T1D, but not Control, mice significantly impaired cognitive performance, and this was associated with hippocampal oxidative damage and inflammation despite an enhanced expression of Nrf2 and its target genes Hmox1 and Nqo1 In Nrf2(-/-) mice, both T1D and Hypo independently resulted in impaired cognitive performance, and this was associated with oxidative cell damage and marked inflammation. Together, these data suggest that Hypo induces an Nrf2-dependent antioxidant response in the hippocampus, which counteracts oxidative damage. However, in T1D, this neuroprotective mechanism is insufficient to prevent neuronal oxidative damage, resulting in chronic deficits in working and long-term memory.
Collapse
Affiliation(s)
- Alison D McNeilly
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, Dundee, U.K
| | - Jennifer R Gallagher
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, Dundee, U.K
| | - Albena T Dinkova-Kostova
- Division of Cancer Research, School of Medicine, Ninewells Hospital and Medical School, Dundee, U.K
| | - John D Hayes
- Division of Cancer Research, School of Medicine, Ninewells Hospital and Medical School, Dundee, U.K
| | - John Sharkey
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, Dundee, U.K. Division of Neuroscience, School of Medicine, Ninewells Hospital and Medical School, Dundee, U.K
| | - Michael L J Ashford
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, Dundee, U.K
| | - Rory J McCrimmon
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital and Medical School, Dundee, U.K.
| |
Collapse
|
34
|
Quincozes-Santos A, Bobermin LD, de Assis AM, Gonçalves CA, Souza DO. Fluctuations in glucose levels induce glial toxicity with glutamatergic, oxidative and inflammatory implications. Biochim Biophys Acta Mol Basis Dis 2016; 1863:1-14. [PMID: 27663722 DOI: 10.1016/j.bbadis.2016.09.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 08/21/2016] [Accepted: 09/17/2016] [Indexed: 02/06/2023]
Abstract
Astrocytes are dynamic cells that maintain brain homeostasis by regulating neurotransmitter systems, antioxidant defenses, inflammatory responses and energy metabolism. Astroglial cells are also primarily responsible for the uptake and metabolism of glucose in the brain. Diabetes mellitus (DM) is a pathological condition characterized by hyperglycemia and is associated with several changes in the central nervous system (CNS), including alterations in glial function. Classically, excessive glucose concentrations are used to induce experimental models of astrocyte dysfunction; however, hypoglycemic episodes may also cause several brain injuries. The main focus of the present study was to evaluate how fluctuations in glucose levels induce cytotoxicity. The culture medium of astroglial cells was replaced twice as follows: (1) from 6mM (control) to 12mM (high glucose), and (2) from 12mM to 0mM (glucose deprivation). Cell viability, mitochondrial function, oxidative/nitrosative stress, glutamate metabolism, inflammatory responses, nuclear factor κB (NFκB) transcriptional activity and p38 mitogen-activated protein kinase (p38 MAPK) levels were assessed. Our in vitro experimental model showed that up and down fluctuations in glucose levels decreased cell proliferation, induced mitochondrial dysfunction, increased oxidative/nitrosative stress with consequent cellular biomolecular damage, impaired glutamate metabolism and increased pro-inflammatory cytokine release. Additionally, activation of the NFκB and p38 signaling pathways were putative mechanisms of the effects of glucose fluctuations on astroglial cells. In summary, for the first time, we show that changes in glucose concentrations, from high-glucose levels to glucose deprivation, exacerbate glial injury.
Collapse
Affiliation(s)
- André Quincozes-Santos
- Departamento de Bioquímica, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Larissa Daniele Bobermin
- Departamento de Bioquímica, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Adriano M de Assis
- Departamento de Bioquímica, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carlos-Alberto Gonçalves
- Departamento de Bioquímica, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Diogo Onofre Souza
- Departamento de Bioquímica, Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
35
|
Impact of glucose fluctuation on acute cerebral infarction in type 2 diabetes. Can J Neurol Sci 2016; 41:486-92. [PMID: 24878474 DOI: 10.1017/s0317167100018539] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE This study aimed to describe the frequency and temporal profile of acute cerebral infarction (ACI) using a continuous glucose monitoring system (CGMS) in patients with and without type 2 diabetes mellitus (T2DM) and explore the impact of blood glucose fluctuations on the short-term prognosis of ACI. METHODS The subjects were divided into four groups: T2DM with acute cerebral infarction (DMCI, Group A, n=56); T2DM without acute cerebral infarction (DM-NCI, Group B, n=36); Acute cerebral infarction patients without T2DM (NDM-CI, Group C, n=54); Healthy control group (NG, Group D, n=36). The National Institutes of Health Stoke Scale (NIHSS) and modified Rankin scale (mRs) were collected in Group A and C. All subjects were monitored for 72 hours using the CGMS. Indices such as fasting blood glucose (FBG) and mean amplitude of glycemic excursions (MAGE) were calculated. Glycemic excursions were compared between Group A, B, C and Group D, respectively. Multiple linear regression analysis and logistic analysis was applied. RESULTS MAGE is related to NIHSS, homocysteine (HCY), HOMA-IR, FBG, CRP and IMT, while NIHSS is related to CRP, HCY, HOMA-IR, IMT. The factors impacting the short-term prognosis of ACI were NIHSS, HBA1C and MAGE. CONCLUSION Larger glucose fluctuations are associated with more stroke risk factors and are associated with a poorer short-term prognosis. More attention should be paid to glucose fluctuations in patients with ACI and a history of T2DM.
Collapse
|
36
|
Abstract
The review considers the current views on the central nervous system (CNS) in diabetes mellitus (DM). It discusses an attitude towards the term 'diabetic encephalopathy'. The data of investigations of cognitive functions in types 1 and 2 DM and the brain structural imaging results obtained using up-to-date technologies are considered. The results of studies of the factors that induce cerebral changes in DM and their associated cognitive impairments are given. There is evidence that hyperglycemia has a more considerable impact on the above processes than hypoglycemia; other possible factors, apart from blood glucose control, are set out. The current views on the function of insulin in the CNS and the relationship of central insulin resistance to Alzheimer's disease are outlined. There are current data on intranasal insulin application that is still exploratory, but, as might be judged from the findings, may by a promising method for the treatment and prevention of cognitive decline in both patients with DM and those without this condition.
Collapse
Affiliation(s)
- E V Surkova
- Endocrinology Research Center, Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
37
|
Prasad SN, Bharath MMS. Neurorestorative effects of eugenol, a spice bioactive: Evidence in cell model and its efficacy as an intervention molecule to abrogate brain oxidative dysfunctions in the streptozotocin diabetic rat. Neurochem Int 2015; 95:24-36. [PMID: 26519099 DOI: 10.1016/j.neuint.2015.10.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Revised: 10/09/2015] [Accepted: 10/24/2015] [Indexed: 02/07/2023]
Abstract
Eugenol (EU), an active principle of cloves, is also widely distributed in various other plants (eg. basil, cinnamon, etc). While its antioxidant and anti-inflammatory properties are well established, biochemical insights related to its neuromodulatory potential in diabetic conditions are not clear. In the present study, initially we investigated its potential to modulate specific biochemical responses in SHSY5Y cells under experimentally -induced hyperglycemic condition. Co-exposure of cells with EU (5-10 μM) not only enhanced the cell viability, but significantly offset glucose -associated oxidative stress (as evidenced by diminished levels of reactive oxygen species and hydroperoxides). Further EU enhanced the reduced glutathione (GSH) levels and also ameliorated the levels of 3 - nitrotyrosine and expression of HSP70. We subsequently examined its efficacy to attenuate biochemical aberrations in brain regions of a streptozotocin (STZ) diabetic rat employing an intervention approach. Brain regions of EU treated (10 mg/kg bw/d, post 6 weeks of STZ) diabetic rats showed diminished levels of oxidative markers and protein carbonyls in both cytosolic and mitochondrial fractions. EU treatment caused enhanced activities of enzymic antioxidants and diminished both GSH and total thiols. Further, activities of complex I - III, succinate dehydrogenase and citrate synthase in brain regions were also significantly restored. Interestingly, EU treatment differentially attenuated the elevated activity of acetylcholinesterase and levels of calcium in brain regions. Collectively, based on the data obtained in in vitro and in vivo models, we hypothesize that EU may be employed as an adjuvant therapeutic molecule to alleviate complications under diabetic conditions.
Collapse
Affiliation(s)
- Sathya N Prasad
- Department of Biochemistry & Nutrition, CSIR - Central Food Technological Research Institute (CFTRI), Mysuru 570020, India
| | - M M Srinivas Bharath
- Department of Neurochemistry, National Institute of Mental Health and Neurosciences (NIMHANS), #2900, Hosur Road, Bengaluru 560029, India
| |
Collapse
|
38
|
Abstract
A constant supply of glucose to the brain is critical for normal cerebral metabolism. The dysglycemia of type 1 diabetes (T1D) can affect activity, survival, and function of neural cells. Clinical studies in T1D have shown impairments in brain morphology and function. The most neurotoxic milieu seems to be young age and/or diabetic ketoacidosis at onset, severe hypoglycemia under the age of 6 years followed by chronic hyperglycemia. Adverse cognitive outcomes seem to be associated with poorer mental health outcomes. It is imperative to improve outcomes by investigating the mechanisms of injury so that neuroprotective strategies independent of glycemia can be identified.
Collapse
Affiliation(s)
- Fergus J Cameron
- Department of Endocrinology and Diabetes, Royal Children's Hospital, Murdoch Childrens Research Institute, 50 Flemington Road, Parkville, Melbourne 3052, Australia; Department of Paediatrics, University of Melbourne, Melbourne 3010, Australia.
| |
Collapse
|
39
|
Sajja RK, Cucullo L. Altered glycaemia differentially modulates efflux transporter expression and activity in hCMEC/D3 cell line. Neurosci Lett 2015; 598:59-65. [PMID: 25982326 DOI: 10.1016/j.neulet.2015.05.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 04/18/2015] [Accepted: 05/09/2015] [Indexed: 11/29/2022]
Abstract
The unique phenotype of blood-brain barrier (BBB) endothelium is partly maintained by abundant expression of ATP-binding cassette superfamily of efflux transporters that strictly restrict the CNS access to toxic substances including xenobiotics in circulation. Previously, we have shown that diabetes-related altered glycemic conditions differentially affect and compromise BBB integrity. However, the impact of diabetes on BBB efflux transporters is less understood. In this study, we examined the effects of single or repeated episodes of hypo-and hyperglycemia on major BBB efflux transporters expression/function in human cerebromicrovascular endothelial cell line (hCMEC/D3). Cells were exposed to normal (5.5 mM), hypo (2.2 mM) or hyper (25 or 35 mM)-glycemic media containing D-glucose for 12h (acute) or two 3h episodes/day of hypo- or hyperglycemia with an intercalated 2h normalglycemic exposure for 3 days ("glycemic variability", see Methods). Acute hypoglycemic exposure (12h) up-regulated BBB endothelial mRNA and protein expression of P-glycoprotein, BCRP and other multidrug resistance associated proteins (MRP1 and 4) paralleled by an increase in transporter-specific efflux activity (∼ 2-fold vs. control). Although, 12h hyperglycemia did not affect the efflux transporter expression (except for MRP4), a significant increase in BCRP activity was observed. By contrast, DNA microarray data revealed that repeated hyperglycemic episodes (but not hypoglycemia) significantly up-regulate P-glycoprotein expression and activity. Thus, this study suggests a differential impact of altered glycemic conditions on major BBB drug efflux transporters expression/function, sensitive to the length of exposure (acute vs. repeated), with an implication for altered CNS drug disposition in diabetic population.
Collapse
Affiliation(s)
- Ravi K Sajja
- Center for Blood-Brain Barrier Research, Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Luca Cucullo
- Center for Blood-Brain Barrier Research, Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| |
Collapse
|
40
|
Biswas J, Goswami P, Gupta S, Joshi N, Nath C, Singh S. Streptozotocin Induced Neurotoxicity Involves Alzheimer's Related Pathological Markers: a Study on N2A Cells. Mol Neurobiol 2015; 53:2794-2806. [PMID: 25823512 DOI: 10.1007/s12035-015-9144-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 03/18/2015] [Indexed: 01/21/2023]
Abstract
Intracerebroventricular (icv) injection of streptozotocin (STZ) in rat brain causes prolonged impairment of brain energy metabolism and oxidative damage and leads to cognitive dysfunction; however, its mechanistic specific effects on neurons are not known. The present study was conducted to investigate the STZ-induced cellular and molecular alterations in mouse neuronal N2A cells. The N2A cells were treated with STZ (10, 50, 100, 1000 μM) for 48 h, and different assays were performed. STZ treatment caused significant decrease in cell viability, choline levels, increased acetylcholinesterase (AChE) activity, tau phosphorylation and amyloid aggregation. STZ treatment also led to low levels of glucose uptake, elevated mitochondrial stress, translocation of cytochrome c in cytosol, phosphatidylserine externalization, increased expression of caspase-3 and DNA damage. Co-treatment of clinically used drug donepezil (1 μM) offered significant protection against STZ induced neurotoxicity. Donepezil treatment significantly inhibited the STZ induced neurotoxicity, altered choline level, AChE activity, lowered glucose uptake and mitochondrial stress. However, the caspase-3 expression remains unaltered with co-treatment of donepezil. In conclusion, findings showed that STZ treated N2A cells exhibited the Alzheimer's disease (AD) related pathological markers which are attenuated with co-treatment of donepezil. Findings of the study suggested the potent use of STZ treated N2A cells as in vitro experimental test model to study the disease mechanism at cellular level.
Collapse
Affiliation(s)
- Joyshree Biswas
- Toxicology Division, Central Drug Research Institute (CSIR-CDRI), Lucknow, UP, 226031, India
| | - Poonam Goswami
- Toxicology Division, Central Drug Research Institute (CSIR-CDRI), Lucknow, UP, 226031, India
| | - Sonam Gupta
- Toxicology Division, Central Drug Research Institute (CSIR-CDRI), Lucknow, UP, 226031, India
| | - Neeraj Joshi
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Chandishwar Nath
- Toxicology Division, Central Drug Research Institute (CSIR-CDRI), Lucknow, UP, 226031, India
| | - Sarika Singh
- Toxicology Division, Central Drug Research Institute (CSIR-CDRI), Lucknow, UP, 226031, India.
| |
Collapse
|
41
|
Lin A, Northam EA, Werther GA, Cameron FJ. Risk factors for decline in IQ in youth with type 1 diabetes over the 12 years from diagnosis/illness onset. Diabetes Care 2015; 38:236-42. [PMID: 25488913 DOI: 10.2337/dc14-1385] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE This study examined illness-related change in intelligence quotient (IQ) in a cohort of youth with type 1 diabetes studied prospectively from disease onset in childhood to follow-up 12 years later in late adolescence/early adulthood. RESEARCH DESIGN AND METHODS Participants included type 1 diabetes patients (n = 95; mean age at follow-up 21.3 years) and healthy control participants (HCs; n = 67; mean age at follow-up 21.0 years) from a cohort followed prospectively. Measures included Wechsler Preschool and Primary Scale of Intelligence-Revised, Wechsler Intelligence Scale for Children-Revised, and Wechsler Abbreviated Scale of Intelligence and prospective collection of data on metabolic control history. RESULTS Young people with type 1 diabetes showed greater decline in verbal IQ (VIQ) and full-scale IQ (FSIQ), but not performance IQ (PIQ), than HCs. Within the diabetes group, a younger age at diabetes onset was associated with a decline in PIQ and FSIQ (P ≤ 0.001). A history of hypoglycemic seizures was associated with a decline in VIQ (P = 0.002). Long-term metabolic control was not associated with changes in IQ. Interaction terms were not significant, suggesting no moderating effect of one diabetes-related variable over another. CONCLUSIONS The presence of diabetes may negatively influence some aspects of IQ over time. Specific illness risk factors, such as an earlier age of disease onset and a history of hypoglycemic seizures, appear to put the young person at greater risk. Academic progress of children identified as at risk should be monitored and educational supports provided if necessary.
Collapse
Affiliation(s)
- Ashleigh Lin
- Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Elisabeth A Northam
- Department of Psychology, Royal Children's Hospital, Melbourne, Australia Department of Endocrinology and Diabetes, Royal Children's Hospital, Melbourne, Australia Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Australia
| | - George A Werther
- Department of Endocrinology and Diabetes, Royal Children's Hospital, Melbourne, Australia Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Australia Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Melbourne, Australia
| | - Fergus J Cameron
- Department of Endocrinology and Diabetes, Royal Children's Hospital, Melbourne, Australia Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Australia Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Melbourne, Australia
| |
Collapse
|
42
|
Rewers MJ, Pillay K, de Beaufort C, Craig ME, Hanas R, Acerini CL, Maahs DM. ISPAD Clinical Practice Consensus Guidelines 2014. Assessment and monitoring of glycemic control in children and adolescents with diabetes. Pediatr Diabetes 2014; 15 Suppl 20:102-14. [PMID: 25182311 DOI: 10.1111/pedi.12190] [Citation(s) in RCA: 226] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 06/16/2014] [Indexed: 12/24/2022] Open
Affiliation(s)
- Marian J Rewers
- Barbara Davis Center, University of Colorado Denver, Aurora, CO, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Gejl M, Rungby J, Brock B, Gjedde A. At the centennial of Michaelis and Menten, competing Michaelis-Menten steps explain effect of GLP-1 on blood-brain transfer and metabolism of glucose. Basic Clin Pharmacol Toxicol 2014; 115:162-71. [PMID: 24684709 DOI: 10.1111/bcpt.12240] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 03/17/2014] [Indexed: 12/16/2022]
Abstract
Glucagon-like peptide-1 (GLP-1) is a potent insulinotropic incretin hormone with both pancreatic and extrapancreatic effects. Studies of GLP-1 reveal significant effects in regions of brain tissue that regulate appetite and satiety. GLP-1 mimetics are used for the treatment of type 2 diabetes mellitus. GLP-1 interacts with peripheral functions in which the autonomic nervous system plays an important role, and emerging pre-clinical findings indicate a potential neuroprotective role of the peptide, for example in models of stroke and in neurodegenerative disorders. A century ago, Leonor Michaelis and Maud Menten described the steady-state enzyme kinetics that still apply to the multiple receptors, transporters and enzymes that define the biochemical reactions of the brain, including the glucose-dependent impact of GLP-1 on blood-brain glucose transfer and metabolism. This MiniReview examines the potential of GLP-1 as a molecule of interest for the understanding of brain energy metabolism and with reference to the impact on brain metabolism related to appetite and satiety regulation, stroke and neurodegenerative disorders. These effects can be understood only by reference to the original formulation of the Michaelis-Menten equation as applied to a chain of kinetically controlled steps. Indeed, the effects of GLP-1 receptor activation on blood-brain glucose transfer and brain metabolism of glucose depend on the glucose concentration and relative affinities of the steps both in vitro and in vivo, as in the pancreas.
Collapse
Affiliation(s)
- Michael Gejl
- Department of Biomedicine - Pharmacology, Aarhus University, Aarhus, Denmark; Centre for Advanced Imaging, The University of Queensland, Brisbane, Qld, Australia
| | | | | | | |
Collapse
|
44
|
Arbelaez AM, Semenkovich K, Hershey T. Glycemic extremes in youth with T1DM: the structural and functional integrity of the developing brain. Pediatr Diabetes 2013; 14:541-53. [PMID: 24119040 PMCID: PMC3857606 DOI: 10.1111/pedi.12088] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 08/14/2013] [Accepted: 09/04/2013] [Indexed: 12/13/2022] Open
Abstract
The adult brain accounts for a disproportionally large percentage of the body’s total energy consumption (1). However, during brain development,energy demand is even higher, reaching the adult rate by age 2 and increasing to nearly twice the adult rate by age 10, followed by gradual reduction toward adult levels in the next decade (1,2). The dramatic changes in brain metabolism occurring over the first two decades of life coincide with the initial proliferation and then pruning of synapses to adult levels.The brain derives its energy almost exclusively from glucose and is largely driven by neuronal signaling, biosynthesis, and neuroprotection (3–6).Glucose homeostasis in the body is tightly regulated by a series of hormones and physiologic responses. As a result, hypoglycemia and hyperglycemia are rare occurrences in normal individuals, but they occur commonly inpatients with type 1 diabetes mellitus (T1DM) due to a dysfunction of peripheral glucose-insulin-glucagon responses and non-physiologic doses of exogenous insulin, which imperfectly mimic normal physiology. These extremes can occur more frequently in children and adolescents with T1DM due to the inadequacies of insulin replacement therapy, events leading to the diagnosis [prolonged untreated hyperglycemia and diabetic ketoacidosis (DKA)], and to behavioral factors interfering with optimal treatment. When faced with fluctuations in glucose supply the metabolism of the body and brain change dramatically, largely to conserve resources and, at a cost to other organs, to preserve brain function (7). However,if the normal physiological mechanisms that prevent these severe glucose fluctuations and maintain homeostasis are impaired, neuronal function and potentially viability can be affected (8–11).
Collapse
Affiliation(s)
- Ana Maria Arbelaez
- Department of Pediatrics, Washington University School of Medicine St. Louis, Missouri, United States, 63110
| | - Katherine Semenkovich
- Department of Pediatrics, Washington University School of Medicine St. Louis, Missouri, United States, 63110
| | - Tamara Hershey
- Department of Psychiatry, Washington University School of Medicine St. Louis, Missouri, United States, 63110,Department of Neurology, Washington University School of Medicine St. Louis, Missouri, United States, 63110,Department of Radiology, Washington University School of Medicine St. Louis, Missouri, United States, 63110
| |
Collapse
|
45
|
Cong WN, Wang R, Cai H, Daimon CM, Scheibye-Knudsen M, Bohr VA, Turkin R, Wood WH, Becker KG, Moaddel R, Maudsley S, Martin B. Long-term artificial sweetener acesulfame potassium treatment alters neurometabolic functions in C57BL/6J mice. PLoS One 2013; 8:e70257. [PMID: 23950916 PMCID: PMC3737213 DOI: 10.1371/journal.pone.0070257] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 06/18/2013] [Indexed: 12/22/2022] Open
Abstract
With the prevalence of obesity, artificial, non-nutritive sweeteners have been widely used as dietary supplements that provide sweet taste without excessive caloric load. In order to better understand the overall actions of artificial sweeteners, especially when they are chronically used, we investigated the peripheral and central nervous system effects of protracted exposure to a widely used artificial sweetener, acesulfame K (ACK). We found that extended ACK exposure (40 weeks) in normal C57BL/6J mice demonstrated a moderate and limited influence on metabolic homeostasis, including altering fasting insulin and leptin levels, pancreatic islet size and lipid levels, without affecting insulin sensitivity and bodyweight. Interestingly, impaired cognitive memory functions (evaluated by Morris Water Maze and Novel Objective Preference tests) were found in ACK-treated C57BL/6J mice, while no differences in motor function and anxiety levels were detected. The generation of an ACK-induced neurological phenotype was associated with metabolic dysregulation (glycolysis inhibition and functional ATP depletion) and neurosynaptic abnormalities (dysregulation of TrkB-mediated BDNF and Akt/Erk-mediated cell growth/survival pathway) in hippocampal neurons. Our data suggest that chronic use of ACK could affect cognitive functions, potentially via altering neuro-metabolic functions in male C57BL/6J mice.
Collapse
Affiliation(s)
- Wei-na Cong
- Metabolism Unit, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Rui Wang
- Metabolism Unit, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Huan Cai
- Metabolism Unit, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Caitlin M. Daimon
- Metabolism Unit, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Morten Scheibye-Knudsen
- Section on DNA repair, Laboratory of Molecular Gerontology, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Vilhelm A. Bohr
- Section on DNA repair, Laboratory of Molecular Gerontology, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Rebecca Turkin
- Metabolism Unit, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, Maryland, United States of America
| | - William H. Wood
- Gene Expression and Genomics Unit, Laboratory of Genetics, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Kevin G. Becker
- Gene Expression and Genomics Unit, Laboratory of Genetics, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Ruin Moaddel
- Bioanalytical Chemistry and Drug Discovery Section, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Stuart Maudsley
- Receptor Pharmacology Unit, Laboratory of Neurosciences, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Bronwen Martin
- Metabolism Unit, Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, Maryland, United States of America
| |
Collapse
|
46
|
|
47
|
Cardoso S, Santos RX, Correia SC, Carvalho C, Santos MS, Baldeiras I, Oliveira CR, Moreira PI. Insulin-induced recurrent hypoglycemia exacerbates diabetic brain mitochondrial dysfunction and oxidative imbalance. Neurobiol Dis 2012; 49:1-12. [PMID: 22940631 DOI: 10.1016/j.nbd.2012.08.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 07/21/2012] [Accepted: 08/16/2012] [Indexed: 12/17/2022] Open
Abstract
Intensive insulin therapy can prevent or slow the progression of long-term diabetes complications but, at the same time, it increases the risk for episodes of severe hypoglycemia. In our study, we used a protocol intended to mimic the levels of blood glucose that occur in type 1 diabetic patients under an intensive insulin therapy. Streptozotocin (STZ)-induced diabetic rats were treated subcutaneously with twice-daily insulin injections for 2weeks to induce hypoglycemic episodes. Brain cortical and hippocampal mitochondria were isolated and mitochondrial bioenergetics (respiratory chain and phosphorylation system) and oxidative status parameters (malondialdehyde (MDA) levels, mitochondrial aconitase activity and enzymatic and non-enzymatic antioxidant defenses) were analyzed. The protein levels of synaptophysin, a marker of synaptic integrity, and caspase 9 activity were also evaluated in cortical and hippocampal homogenates. Brain cortical mitochondria isolated from hyper- and recurrent hypoglycemic animals presented higher levels of MDA and α-tocopherol together with an increased glutathione disulfide reductase activity, lower manganese superoxide dismutase (MnSOD) activity and glutathione-to-glutathione disulfide (GSH/GSSG) ratio. No significant alterations were found in cortical mitochondrial respiratory chain and oxidative phosphorylation system. Hippocampal mitochondria from both experimental groups presented an impaired oxidative phosphorylation system characterized by a decreased mitochondrial energization potential and ATP levels and higher repolarization lag phase. In addition, higher MDA levels and decreased GSH/GSSG, α-tocopherol levels, and aconitase, glutathione peroxidase and MnSOD activities were observed in both groups of animals. Hippocampal mitochondria from recurrent hypoglycemic animals also showed an impairment of the respiratory chain characterized by a lower state 3 of respiration, respiratory control ratio and ADP/O index, and a higher state 4 of respiration. Additionally, a non-statistically significant decrease in synaptophysin protein levels was observed in cortical homogenates from recurrent hypoglycemic rats as well as in hippocampal homogenates from hyperglycemic and recurrent hypoglycemic rats. An increase in caspase 9 activity was also observed in hippocampal homogenates from hyperglycemic and recurrent hypoglycemic animals. Our results show that mitochondrial dysfunction induced by long-term hyperglycemic effects is exacerbated by recurrent hypoglycemia, which may compromise the function and integrity of brain cells.
Collapse
Affiliation(s)
- Susana Cardoso
- Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Portugal
| | - Renato X Santos
- Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Portugal
| | - Sónia C Correia
- Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Portugal
| | - Cristina Carvalho
- Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Portugal
| | - Maria S Santos
- Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Portugal
| | - Inês Baldeiras
- Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Laboratory of Neurochemistry, Coimbra University Hospital, Portugal; Neurology Department, Faculty of Medicine, University of Coimbra, Portugal
| | - Catarina R Oliveira
- Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Institute of Biochemistry, Faculty of Medicine, University of Coimbra, Portugal
| | - Paula I Moreira
- Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-354 Coimbra, Portugal.
| |
Collapse
|