1
|
Sanchez-Rangel E, Gunawan F, Jiang L, Savoye M, Dai F, Coppoli A, Rothman DL, Mason GF, Hwang JJ. Reversibility of brain glucose kinetics in type 2 diabetes mellitus. Diabetologia 2022; 65:895-905. [PMID: 35247067 PMCID: PMC8960594 DOI: 10.1007/s00125-022-05664-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 12/02/2021] [Indexed: 11/20/2022]
Abstract
AIMS/HYPOTHESIS We have previously shown that individuals with uncontrolled type 2 diabetes have a blunted rise in brain glucose levels measured by 1H magnetic resonance spectroscopy. Here, we investigate whether reductions in HbA1c normalise intracerebral glucose levels. METHODS Eight individuals (two men, six women) with poorly controlled type 2 diabetes and mean ± SD age 44.8 ± 8.3 years, BMI 31.4 ± 6.1 kg/m2 and HbA1c 84.1 ± 16.2 mmol/mol (9.8 ± 1.4%) underwent 1H MRS scanning at 4 Tesla during a hyperglycaemic clamp (~12.21 mmol/l) to measure changes in cerebral glucose at baseline and after a 12 week intervention that improved glycaemic control through the use of continuous glucose monitoring, diabetes regimen intensification and frequent visits to an endocrinologist and nutritionist. RESULTS Following the intervention, mean ± SD HbA1c decreased by 24.3 ± 15.3 mmol/mol (2.1 ± 1.5%) (p=0.006), with minimal weight changes (p=0.242). Using a linear mixed-effects regression model to compare glucose time courses during the clamp pre and post intervention, the pre-intervention brain glucose level during the hyperglycaemic clamp was significantly lower than the post-intervention brain glucose (p<0.001) despite plasma glucose levels during the hyperglycaemic clamp being similar (p=0.266). Furthermore, the increases in brain glucose were correlated with the magnitude of improvement in HbA1c (r = 0.71, p=0.048). CONCLUSION/INTERPRETATION These findings highlight the potential reversibility of cerebral glucose transport capacity and metabolism that can occur in individuals with type 2 diabetes following improvement of glycaemic control. Trial registration ClinicalTrials.gov NCT03469492.
Collapse
Affiliation(s)
- Elizabeth Sanchez-Rangel
- Department of Internal Medicine/Section of Endocrinology, Yale University School of Medicine, New Haven, CT, USA
| | - Felona Gunawan
- Department of Internal Medicine/Section of Endocrinology, Yale University School of Medicine, New Haven, CT, USA
| | - Lihong Jiang
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Mary Savoye
- Department of Pediatric Endocrinology and General Clinical Research Center, Yale University School of Medicine, New Haven, CT, USA
| | - Feng Dai
- Yale Center for Analytical Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Anastasia Coppoli
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Douglas L Rothman
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University School of Medicine, New Haven, CT, USA
| | - Graeme F Mason
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Janice Jin Hwang
- Department of Internal Medicine/Section of Endocrinology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
2
|
Chen Z, Guo H, Lu Z, Sun K, Jin Q. Hyperglycemia aggravates spinal cord injury through endoplasmic reticulum stress mediated neuronal apoptosis, gliosis and activation. Biomed Pharmacother 2019; 112:108672. [PMID: 30784940 DOI: 10.1016/j.biopha.2019.108672] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 01/29/2019] [Accepted: 02/05/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Hyperglycemia has been shown to influence prognostic outcome of spinal cord injury (SCI). However, the corresponding mechanism is not very clear. AIM This study is expected to explore the role of endoplasmic reticulum (ER) stress in hyperglycemia aggravated SCI. METHODS Hyperglycemia was established in rats by intraperitoneal (i.p.) injection of streptozotocin. SCI was performed at the T10 of spinal cord through weight dropping. ER stress was suppressed by oral gavage of 4-PBA. ER stress, histological change of the injured spinal cords, neuronal apoptosis, demyelination, glial proliferation, inflammatory factor production, blood-spinal cord barrier (BSCB) permeability, TJ (Occludin, Claudin5) and AJ (β-catenin, P120) protein degradation, and locomotor recovery were determined using western blotting, immunohistochemistry, HE staining, Evan's Blue assay, BBB scores and inclined plane test, respectively. In vitro, rat spinal cord neurons cells (RSCNCs) and cerebral microvascular endothelial cells (RCMECs) were stimulated with high glucose (HG) and/or thapsigargin (TG). The effects of HG and/or TG on RSCNCs apoptosis, and AJ and TJ expression by RCMECs were evaluated with flow cytometry and western blotting, respectively. RESULTS Hyperglycemic rats exhibited enhanced ER stress, increased neuronal apoptosis, aggravated demyelination, increased glial proliferation and inflammatory factors secretion, more serious BSCB disruption and disturbed locomotor recovery. ER stress inhibition alleviated hyperglycemia induced adverse effect on neuronal apoptosis and BSCB permeability, whereas showed little influence on glial activation and inflammation. CONCLUSION ER stress was aggravated in hyperglycemic rats after SCI, and subsequently promoted neuronal apoptosis and BSCB disruption in rats.
Collapse
Affiliation(s)
- Zhirong Chen
- Department of Orthopedics, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Haohui Guo
- Department of Orthopedics, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Zhidong Lu
- Department of Orthopedics, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Kening Sun
- Department of Orthopedics, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Qunhua Jin
- Department of Orthopedics, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
3
|
Duarte JMN, Skoug C, Silva HB, Carvalho RA, Gruetter R, Cunha RA. Impact of Caffeine Consumption on Type 2 Diabetes-Induced Spatial Memory Impairment and Neurochemical Alterations in the Hippocampus. Front Neurosci 2019; 12:1015. [PMID: 30686981 PMCID: PMC6333904 DOI: 10.3389/fnins.2018.01015] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 12/17/2018] [Indexed: 12/20/2022] Open
Abstract
Diabetes affects the morphology and plasticity of the hippocampus, and leads to learning and memory deficits. Caffeine has been proposed to prevent memory impairment upon multiple chronic disorders with neurological involvement. We tested whether long-term caffeine consumption prevents type 2 diabetes (T2D)-induced spatial memory impairment and hippocampal alterations, including synaptic degeneration, astrogliosis, and metabolic modifications. Control Wistar rats and Goto-Kakizaki (GK) rats that develop T2D were treated with caffeine (1 g/L in drinking water) for 4 months. Spatial memory was evaluated in a Y-maze. Hippocampal metabolic profile and glucose homeostasis were investigated by 1H magnetic resonance spectroscopy. The density of neuronal, synaptic, and glial-specific markers was evaluated by Western blot analysis. GK rats displayed reduced Y-maze spontaneous alternation and a lower amplitude of hippocampal long-term potentiation when compared to controls, suggesting impaired hippocampal-dependent spatial memory. Diabetes did not impact the relation of hippocampal to plasma glucose concentrations, but altered the neurochemical profile of the hippocampus, such as increased in levels of the osmolites taurine (P < 0.001) and myo-inositol (P < 0.05). The diabetic hippocampus showed decreased density of the presynaptic proteins synaptophysin (P < 0.05) and SNAP25 (P < 0.05), suggesting synaptic degeneration, and increased GFAP (P < 0.001) and vimentin (P < 0.05) immunoreactivities that are indicative of astrogliosis. The effects of caffeine intake on hippocampal metabolism added to those of T2D, namely reducing myo-inositol levels (P < 0.001) and further increasing taurine levels (P < 0.05). Caffeine prevented T2D-induced alterations of GFAP, vimentin and SNAP25, and improved memory deficits. We conclude that caffeine consumption has beneficial effects counteracting alterations in the hippocampus of GK rats, leading to the improvement of T2D-associated memory impairment.
Collapse
Affiliation(s)
- João M N Duarte
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Laboratory for Functional and Metabolic Imaging, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Cecilia Skoug
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Henrique B Silva
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Rui A Carvalho
- Department of Life Sciences, Faculty of Science and Technology, University of Coimbra, Coimbra, Portugal
| | - Rolf Gruetter
- Laboratory for Functional and Metabolic Imaging, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Department of Radiology, University of Lausanne, Lausanne, Switzerland.,Department of Radiology, University of Geneva, Geneva, Switzerland
| | - Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
4
|
Van Dyken P, Lacoste B. Impact of Metabolic Syndrome on Neuroinflammation and the Blood-Brain Barrier. Front Neurosci 2018; 12:930. [PMID: 30618559 PMCID: PMC6297847 DOI: 10.3389/fnins.2018.00930] [Citation(s) in RCA: 209] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/27/2018] [Indexed: 12/29/2022] Open
Abstract
Metabolic syndrome, which includes diabetes and obesity, is one of the most widespread medical conditions. It induces systemic inflammation, causing far reaching effects on the body that are still being uncovered. Neuropathologies triggered by metabolic syndrome often result from increased permeability of the blood-brain-barrier (BBB). The BBB, a system designed to restrict entry of toxins, immune cells, and pathogens to the brain, is vital for proper neuronal function. Local and systemic inflammation induced by obesity or type 2 diabetes mellitus can cause BBB breakdown, decreased removal of waste, and increased infiltration of immune cells. This leads to disruption of glial and neuronal cells, causing hormonal dysregulation, increased immune sensitivity, or cognitive impairment depending on the affected brain region. Inflammatory effects of metabolic syndrome have been linked to neurodegenerative diseases. In this review, we discuss the effects of obesity and diabetes-induced inflammation on the BBB, the roles played by leptin and insulin resistance, as well as BBB changes occurring at the molecular level. We explore signaling pathways including VEGF, HIFs, PKC, Rho/ROCK, eNOS, and miRNAs. Finally, we discuss the broader implications of neural inflammation, including its connection to Alzheimer's disease, multiple sclerosis, and the gut microbiome.
Collapse
Affiliation(s)
- Peter Van Dyken
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Baptiste Lacoste
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
5
|
Inhibition of Endoplasmic Reticulum Stress Preserves the Integrity of Blood-Spinal Cord Barrier in Diabetic Rats Subjected to Spinal Cord Injury. Sci Rep 2017; 7:7661. [PMID: 28794417 PMCID: PMC5550423 DOI: 10.1038/s41598-017-08052-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 07/04/2017] [Indexed: 12/23/2022] Open
Abstract
The blood-spinal cord barrier (BSCB) plays significance roles in recovery following spinal cord injury (SCI), and diabetes mellitus (DM) impairs endothelial cell function and integrity of BSCS. Endoplasmic reticulum (ER) stress occurs in the early stages of SCI and affects prognosis and cell survival. However, the relationship between ER stress and the integrity of BSCB in diabetic rats after SCI remains unclear. Here we observed that diabetic rats showed increased extravasation of Evans Blue (EB) dye, and loss of endothelial cells and pericytes 1 day after SCI compared to non-diabetic rats. Diabetes was also shown to induce activation of ER stress. Similar effects were observed in human brain microvascular endothelial cells. 4-phenylbutyric acid (4-PBA), an ER stress inhibitor lowered the adverse effect of diabetes on SCI, reduced EB dye extravasation, and limited the loss of endothelial cells and pericytes. Moreover, 4-PBA treatment partially reversed the degradation of tight junction and adherens junction both in vivo and in vitro. In conclusion, diabetes exacerbates the disruption of BSCB after SCI via inducing ER stress, and inhibition of ER stress by 4-PBA may play a beneficial role on the integrity of BSCB in diabetic SCI rats, leading to improved prognosis.
Collapse
|
6
|
Sonneville R, Vanhorebeek I, den Hertog HM, Chrétien F, Annane D, Sharshar T, Van den Berghe G. Critical illness-induced dysglycemia and the brain. Intensive Care Med 2014; 41:192-202. [PMID: 25465908 DOI: 10.1007/s00134-014-3577-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 11/19/2014] [Indexed: 01/04/2023]
Abstract
PURPOSE Dysglycemia is a characteristic feature of critical illness associated with adverse outcome. Whether dysglycemia contributes to brain dysfunction during critical illness and long-term neurological complications is unclear. We give an overview of glucose metabolism in the brain and review the literature on critical illness-induced dysglycemia and the brain. METHODS Medline database search using relevant search terms on dysglycemia, critical illness, acute brain injury/dysfunction, and randomized controlled trial. RESULTS Hyperglycemia has been associated with deleterious effects on the nervous system. Underlying mechanisms in critical illness remain largely speculative and are often extrapolated from knowledge in diabetes mellitus. Increased hyperglycemia-induced blood-brain barrier permeability, oxidative stress, and microglia activation may play a role and compromise neuronal and glial cell integrity. Hypoglycemia is feared as critically ill patients cannot recognize or communicate hypoglycemic symptoms, which furthermore are masked by sedation and analgesia. However, observational data on the impact of brief hypoglycemia on the brain in critical illness are controversial. Secondary analysis of two large randomized studies suggested neuroprotection by strict glycemic control with insulin during intensive care, with lowered intracranial pressure, reduction of seizures, and better long-term rehabilitation in patients with isolated brain injury, and reduced incidence of critical illness polyneuromyopathy in the general critically ill patient population. Several subsequent studies failed to reproduce neurological benefit, likely explained by methodological issues, which include divergent achieved glucose levels and inaccurate glucose monitoring tools. CONCLUSIONS Preventing hyperglycemia during critical illness holds promise as a neuroprotective strategy to preserve brain cell viability and prevent acute brain dysfunction and long-term cognitive impairment in survivors.
Collapse
Affiliation(s)
- Romain Sonneville
- Clinical Department and Laboratory of Intensive Care Medicine, Division of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|
7
|
Rostami E. Glucose and the injured brain-monitored in the neurointensive care unit. Front Neurol 2014; 5:91. [PMID: 24936196 PMCID: PMC4047514 DOI: 10.3389/fneur.2014.00091] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 05/23/2014] [Indexed: 12/16/2022] Open
Abstract
Brain has a continuous demand for energy that is met by oxidative metabolism of oxygen and glucose. This demand is compromised in the injured brain and if the inadequate supply persists it will lead to permanent tissue damage. Zero values of cerebral glucose have been associated with infarction and poor neurological outcome. Furthermore, hyperglycemia is common in patients with neurological insults and associated with poor outcome. Intensive insulin therapy (IIT) to control blood glucose has been suggested and used in neurointensive care with conflicting results. This review covers the studies reporting on monitoring of cerebral glucose with microdialysis in patients with traumatic brain injury (TBI), subarachnoid hemorrhage (SAH) and ischemic stroke. Studies investigating IIT are also discussed. Available data suggest that low cerebral glucose in patients with TBI and SAH provides valuable information on development of secondary ischemia and has been correlated with worse outcome. There is also indication that the location of the catheter is important for correlation between plasma and brain glucose. In conclusion considering catheter location, monitoring of brain glucose in the neurointensive care not only provides information on imminent secondary ischemia it also reveals the effect of peripheral treatment on the injured brain.
Collapse
Affiliation(s)
- Elham Rostami
- Department of Neuroscience, Section of Neurosurgery, Uppsala University , Uppsala , Sweden ; Department of Neuroscience, Karolinska Institutet , Stockholm , Sweden
| |
Collapse
|
8
|
Gulanski BI, De Feyter HM, Page KA, Belfort-DeAguiar R, Mason GF, Rothman DL, Sherwin RS. Increased brain transport and metabolism of acetate in hypoglycemia unawareness. J Clin Endocrinol Metab 2013; 98:3811-20. [PMID: 23796565 PMCID: PMC4425818 DOI: 10.1210/jc.2013-1701] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
CONTEXT Intensive insulin therapy reduces the risk for long-term complications in patients with type 1 diabetes mellitus (T1DM) but increases the risk for hypoglycemia-associated autonomic failure (HAAF), a syndrome that includes hypoglycemia unawareness and defective glucose counterregulation (reduced epinephrine and glucagon responses to hypoglycemia). OBJECTIVE The objective of the study was to address mechanisms underlying HAAF, we investigated whether nonglucose fuels such as acetate, a monocarboxylic acid (MCA), can support cerebral energetics during hypoglycemia in T1DM individuals with hypoglycemia unawareness. DESIGN Magnetic resonance spectroscopy was used to measure brain transport and metabolism of [2-(13)C]acetate under hypoglycemic conditions. SETTING The study was conducted at the Yale Center for Clinical Investigation Hospital Research Unit, Yale Magnetic Resonance Research Center. PATIENTS AND OTHER PARTICIPANTS T1DM participants with moderate to severe hypoglycemia unawareness (n = 7), T1DM controls without hypoglycemia unawareness (n = 5), and healthy nondiabetic controls (n = 10) participated in the study. MAIN OUTCOME MEASURE(S) Brain acetate concentrations, (13)C percent enrichment of glutamine and glutamate, and absolute rates of acetate metabolism were measured. RESULTS Absolute rates of acetate metabolism in the cerebral cortex were 1.5-fold higher among T1DM/unaware participants compared with both control groups during hypoglycemia (P = .001). Epinephrine levels of T1DM/unaware subjects were significantly lower than both control groups (P < .05). Epinephrine levels were inversely correlated with levels of cerebral acetate use across the entire study population (P < .01), suggesting a relationship between up-regulated brain MCA use and HAAF. CONCLUSION Increased MCA transport and metabolism among T1DM individuals with hypoglycemia unawareness may be a mechanism to supply the brain with nonglucose fuels during episodes of acute hypoglycemia and may contribute to the syndrome of hypoglycemia unawareness, independent of diabetes.
Collapse
|
9
|
Abstract
The occurrence of altered brain glucose metabolism has long been suggested in both diabetes and Alzheimer’s diseases. However, the preceding mechanism to altered glucose metabolism has not been well understood. Glucose enters the brain via glucose transporters primarily present at the blood-brain barrier. Any changes in glucose transporter function and expression dramatically affects brain glucose homeostasis and function. In the brains of both diabetic and Alzheimer’s disease patients, changes in glucose transporter function and expression have been observed, but a possible link between the altered glucose transporter function and disease progress is missing. Future recognition of the role of new glucose transporter isoforms in the brain may provide a better understanding of brain glucose metabolism in normal and disease states. Elucidation of clinical pathological mechanisms related to glucose transport and metabolism may provide common links to the etiology of these two diseases. Considering these facts, in this review we provide a current understanding of the vital roles of a variety of glucose transporters in the normal, diabetic and Alzheimer’s disease brain.
Collapse
Affiliation(s)
- Kaushik Shah
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, 1300 S. Coulter Street, Amarillo, TX 79106, USA.
| | | | | |
Collapse
|
10
|
The effects of abnormalities of glucose homeostasis on the expression and binding of muscarinic receptors in cerebral cortex of rats. Eur J Pharmacol 2011; 651:128-36. [DOI: 10.1016/j.ejphar.2010.11.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 11/10/2010] [Accepted: 11/12/2010] [Indexed: 11/23/2022]
|
11
|
Brain glucose overexposure and lack of acute metabolic flexibility in obesity and type 2 diabetes: a PET-[18F]FDG study in Zucker and ZDF rats. J Cereb Blood Flow Metab 2010; 30:895-9. [PMID: 20179723 PMCID: PMC2949188 DOI: 10.1038/jcbfm.2010.27] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Brain glucose exposure may complicate diabetes and obesity. We used positron emission tomography with (18)F-fluorodeoxyglucose in Zucker obese, diabetic, and control rats to determine the contributions of blood glucose mass action versus local mechanisms in regulating central glucose disposal in fasted and acutely glucose-stimulated states, and their adaptations in obesity and diabetes. Our study data indicate that brain glucose uptake is dependent on both local and mass action components, and is stimulated by acute glucose intake in healthy rats. In diseased animals, the organ was chronically overexposed to glucose, due to high fasting glucose uptake, almost abolishing the physiologic response to glucose loading.
Collapse
|
12
|
Duarte JMN, Carvalho RA, Cunha RA, Gruetter R. Caffeine consumption attenuates neurochemical modifications in the hippocampus of streptozotocin-induced diabetic rats. J Neurochem 2009; 111:368-79. [PMID: 19694901 DOI: 10.1111/j.1471-4159.2009.06349.x] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Type 1 diabetes can affect hippocampal function triggering cognitive impairment through unknown mechanisms. Caffeine consumption prevents hippocampal degeneration and memory dysfunction upon different insults and is also known to affect peripheral glucose metabolism. Thus we now characterized glucose transport and the neurochemical profile in the hippocampus of streptozotocin-induced diabetic rats using in vivo(1)H NMR spectroscopy and tested the effect of caffeine consumption thereupon. We found that hippocampal glucose content and transport were unaltered in diabetic rats, irrespective of caffeine consumption. However diabetic rats displayed alterations in their hippocampal neurochemical profile, which were normalized upon restoration of normoglycaemia, with the exception of myo-inositol that remained increased (36 +/- 5%, p < 0.01 compared to controls) likely reflecting osmolarity deregulation. Compared to controls, caffeine-consuming diabetic rats displayed increased hippocampal levels of myo-inositol (15 +/- 5%, p < 0.05) and taurine (23 +/- 4%, p < 0.01), supporting the ability of caffeine to control osmoregulation. Compared to controls, the hippocampus of diabetic rats displayed a reduced density of synaptic proteins syntaxin, synaptophysin and synaptosome-associated protein of 25 kDa (in average 18 +/- 1%, p < 0.05) as well increased glial fibrillary acidic protein (20 +/- 5%, p < 0.05), suggesting synaptic degeneration and astrogliosis, which were prevented by caffeine consumption. In conclusion, neurochemical alterations in the hippocampus of diabetic rats are not related to defects of glucose transport but likely reflect osmoregulatory adaptations caused by hyperglycemia. Furthermore, caffeine consumption affected this neurochemical adaptation to high glucose levels, which may contribute to its potential neuroprotective effects, namely preventing synaptic degeneration and astrogliosis.
Collapse
Affiliation(s)
- João M N Duarte
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
| | | | | | | |
Collapse
|
13
|
Bree AJ, Puente EC, Daphna-Iken D, Fisher SJ. Diabetes increases brain damage caused by severe hypoglycemia. Am J Physiol Endocrinol Metab 2009; 297:E194-201. [PMID: 19435850 PMCID: PMC2711670 DOI: 10.1152/ajpendo.91041.2008] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Insulin-induced severe hypoglycemia causes brain damage. The hypothesis to be tested was that diabetes portends to more extensive brain tissue damage following an episode of severe hypoglycemia. Nine-week-old male streptozotocin-diabetic (DIAB; n = 10) or vehicle-injected control (CONT; n = 7) Sprague-Dawley rats were subjected to hyperinsulinemic (0.2 U.kg(-1).min(-1)) severe hypoglycemic (10-15 mg/dl) clamps while awake and unrestrained. Groups were precisely matched for depth and duration (1 h) of severe hypoglycemia (CONT 11 +/- 0.5 and DIAB 12 +/- 0.2 mg/dl, P = not significant). During severe hypoglycemia, an equal number of episodes of seizure-like activity were noted in both groups. One week later, histological analysis demonstrated extensive neuronal damage in regions of the hippocampus, especially in the dentate gyrus and CA1 regions and less so in the CA3 region (P < 0.05), although total hippocampal damage was not different between groups. However, in the cortex, DIAB rats had significantly (2.3-fold) more dead neurons than CONT rats (P < 0.05). There was a strong correlation between neuronal damage and the occurrence of seizure-like activity (r(2) > 0.9). Separate studies conducted in groups of diabetic (n = 5) and nondiabetic (n = 5) rats not exposed to severe hypoglycemia showed no brain damage. In summary, under the conditions studied, severe hypoglycemia causes brain damage in the cortex and regions within the hippocampus, and the extent of damage is closely correlated to the presence of seizure-like activity in nonanesthetized rats. It is concluded that, in response to insulin-induced severe hypoglycemia, diabetes uniquely increases the vulnerability of specific brain areas to neuronal damage.
Collapse
Affiliation(s)
- Adam J Bree
- Division of Endocrinology, Metabolism, & Lipid Research, Washington University in St. Louis, Campus Box 8127, 660 South Euclid Ave., St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
14
|
Ahmad F, Yusof APM, Bainbridge M, Ab Ghani S. The application of glucose biosensor in studying the effects of insulin and anti-hypertensive drugs towards glucose level in brain striatum. Biosens Bioelectron 2008; 23:1862-8. [PMID: 18440218 DOI: 10.1016/j.bios.2008.03.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2007] [Revised: 03/03/2008] [Accepted: 03/04/2008] [Indexed: 12/20/2022]
Abstract
The mechanisms involving insulin and anti-hypertensive drugs regulation for in vivo cerebral glucose metabolism are not well-understood. This might be due to lack of direct means of measuring cerebral glucose. It is known that the continuous delivery of glucose to the brain is critical for its normal metabolic function. In this study, we report the effect of insulin and anti-hypertensive drugs on glucose level in the striatum of rats. The rats were divided into two groups, i.e. hyperglycemia (14.8+/-0.3mM plasma glucose) and diabetic (10.8+/-0.2mM plasma glucose). A custom-built glucose microsensor was implanted at coordinates A/P 1.0 from bregma, M/L +2.5 and D/V -5.0 (from dura) in the striatum. The amperometric response obtained at +0.23 V vs. Ag|AgCl corresponded to the glucose level in striatum. By varying the concentrations of protaminc zinc insulin infused into the rats, striatum glucose level was found to remain constant throughout, i.e. 9.8+/-0.1 and 4.7+/-0.1mM for hyperglycemic rats and for diabetic rats, respectively. However, infusion of valsartan and felodipine has lowered the striatum glucose level significantly. These findings agreed with the hypothesis that suggested striatum glucose uptake do not depend on insulin but is clearly dependant on anti-hypertensive drugs administration.
Collapse
Affiliation(s)
- Farook Ahmad
- Pusat Pengajian Sains Kimia, Universiti Sains Malaysia, 11800 USM, Pulau Pinang, Malaysia
| | | | | | | |
Collapse
|
15
|
Huang CW, Tsai JJ, Ou HY, Wang ST, Cheng JT, Wu SN, Huang CC. Diabetic hyperglycemia is associated with the severity of epileptic seizures in adults. Epilepsy Res 2008; 79:71-7. [DOI: 10.1016/j.eplepsyres.2007.12.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2007] [Revised: 12/15/2007] [Accepted: 12/29/2007] [Indexed: 10/22/2022]
|
16
|
Puchowicz MA, Xu K, Sun X, Ivy A, Emancipator D, LaManna JC. Diet-induced ketosis increases capillary density without altered blood flow in rat brain. Am J Physiol Endocrinol Metab 2007; 292:E1607-15. [PMID: 17284577 DOI: 10.1152/ajpendo.00512.2006] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It is recognized that ketone bodies, such as R-beta-hydroxybutyrate (beta-HB) and acetoacetate, are energy sources for the brain. As with glucose metabolism, monocarboxylate uptake by the brain is dependent on the function and regulation of its own transporter system. We concurrently investigated ketone body influx, blood flow, and regulation of monocarboxylate transporter (MCT-1) and glucose transporter (GLUT-1) in diet-induced ketotic (KG) rat brain. Regional blood-to-brain beta-HB influx (micromol.g(-1).min(-1)) increased 40-fold with ketosis (4.8 +/- 1.8 plasmabeta-HB; mM) in all regions compared with the nonketotic groups (standard and no-fat diets); there were no changes in regional blood flow. Immunohistochemical staining revealed that GLUT-1 density (number/mm2) in the cortex was significantly elevated (40%) in the ketotic group compared with the standard and no-fat diet groups. MCT-1 was also markedly (3-fold) upregulated in the ketotic group compared with the standard diet group. In the standard diet group, 40% of the brain capillaries stained positive for MCT-1; this amount doubled with the ketotic diet. Western blot analysis of isolated microvessels from ketotic rat brain showed an eightfold increase in GLUT-1 and a threefold increase in MCT-1 compared with the standard diet group. These data suggest that diet-induced ketosis results in increased vascular density at the blood-brain barrier without changes in blood flow. The increase in extraction fraction and capillary density with increased plasma ketone bodies indicates a significant flux of substrates available for brain energy metabolism.
Collapse
Affiliation(s)
- Michelle A Puchowicz
- Department of Anatomy, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106-4938, USA.
| | | | | | | | | | | |
Collapse
|
17
|
Abstract
The specialized cerebral microvascular endothelium interacts with the cellular milieu of the brain and extracellular matrix to form a neurovascular unit, one aspect of which is a regulated interface between the blood and central nervous system (CNS). The concept of this blood-brain barrier (BBB) as a dynamically regulated system rather than a static barrier has wide-ranging implications for pathophysiology of the CNS. While in vitro models of the BBB are useful for screening drugs targeted to the CNS and indispensable for studies of cerebral endothelial cell biology, the complex interactions of the neurovascular unit make animal-based models and methods essential tools for understanding the pathophysiology of the BBB. BBB dysfunction is a complication of neurodegenerative disease and brain injury. Studies on animal models have shown that diseases of the periphery, such as diabetes and inflammatory pain, have deleterious effects on the BBB which may contribute to neurological complications associated with these conditions. Furthermore, genetic and/or epigenetic abnormalities in constituents of the BBB may be significant contributing factors in disease etiology. Research that approaches the BBB as a dynamic system integrated with both the CNS and the periphery is therefore critical to understanding and treating diseases of the CNS. Herein, we review various methodological approaches used to study BBB function in the context of disease. These include measurement of transport between blood and brain, imaging-based technologies, and genomic/proteomic approaches.
Collapse
|
18
|
Mason GF, Petersen KF, Lebon V, Rothman DL, Shulman GI. Increased brain monocarboxylic acid transport and utilization in type 1 diabetes. Diabetes 2006; 55:929-34. [PMID: 16567513 PMCID: PMC2995526 DOI: 10.2337/diabetes.55.04.06.db05-1325] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We hypothesized that increased capacity for brain utilization of nonglucose substrates (monocarboxylic acids [MCAs]) by upregulation of the MCA transporters may contribute metabolic substrates during hypoglycemia. To test this hypothesis, we assessed brain acetate metabolism in five well-controlled type 1 diabetic subjects and six nondiabetic control subjects using 13C magnetic resonance spectroscopy during infusions of [2-(13)C]acetate during hypoglycemia (approximately 55 mg/dl). Acetate is transported into the brain through MCA transporters that are also used for lactate and ketones. Brain acetate concentrations were over twofold higher in the subjects with diabetes than the control subjects (P = 0.01). The fraction of oxidative metabolism from acetate (P = 0.015) and the rate of MCA transport (P = 0.01) were also approximately twofold higher in the diabetic subjects. We conclude that during hypoglycemia MCA transport in the brain was increased by approximately twofold in patients with well-controlled type 1 diabetes, as reflected by higher brain acetate concentrations and rates of acetate oxidation. This upregulation would potentially allow a similar twofold increase in the transport of other MCAs, including lactate, during insulin-induced hypoglycemia. These data are consistent with the hypothesis that upregulation of MCA transport may contribute to the maintenance of brain energetics during hypoglycemia in patients with type 1 diabetes.
Collapse
Affiliation(s)
- Graeme F. Mason
- Department of Psychiatry and Diagnostic Radiology, Yale School of Medicine, New Haven, Connecticut
- Department of Diagnostic Radiology, Yale School of Medicine, New Haven, Connecticut
| | - Kitt F. Petersen
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Vincent Lebon
- Groupe de Spectroscopie RMN, Unité d'Imagerie Isotopique Biochimique et Pharmacologique, Orsay Cedex, France
| | - Douglas L. Rothman
- Department of Diagnostic Radiology, Yale School of Medicine, New Haven, Connecticut
- Department of Biomedical Engineering, Yale School of Medicine, New Haven, Connecticut
| | - Gerald I. Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Department of Cellular and Molecular Physiology, Howard Hughes Medical Institute, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
19
|
YANAI S, OKAICHI Y, OKAICHI H. QUADRATIC RELATIONSHIP BETWEEN LONG-TERM DIETARY RESTRICTION AND SPATIAL COGNITION IN RATS. PSYCHOLOGIA 2006. [DOI: 10.2117/psysoc.2006.214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Shuichi YANAI
- Doshisha University
- Japan Society for the Promotion of Science
| | | | | |
Collapse
|
20
|
Seaquist ER, Tkac I, Damberg G, Thomas W, Gruetter R. Brain glucose concentrations in poorly controlled diabetes mellitus as measured by high-field magnetic resonance spectroscopy. Metabolism 2005; 54:1008-13. [PMID: 16092049 DOI: 10.1016/j.metabol.2005.02.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Hyperglycemia and diabetes alter the function and metabolism of many tissues. The effect on the brain remains poorly defined, but some animal data suggest that chronic hyperglycemia reduces rates of brain glucose transport and/or metabolism. To address this question in human beings, we measured glucose in the occipital cortex of patients with poorly controlled diabetes and healthy volunteers at the same levels of plasma glucose using proton magnetic resonance spectroscopy. Fourteen patients with poorly controlled diabetes (hemoglobin A 1c = 9.8% +/- 1.7%, mean +/- SD) and 14 healthy volunteers similar with respect to age, sex, and body mass index were studied at a plasma glucose of 300 mg/dL. Brain glucose concentrations of patients with poorly controlled diabetes were lower but not statistically different from those of control subjects (4.7 +/- 0.9 vs 5.3 +/- 1.1 micromol/g wet wt; P = .1). Our sample size gave 80% power to detect a difference as small as 1.1 micromol/g wet wt. We conclude that chronic hyperglycemia in diabetes does not alter brain glucose concentrations in human subjects.
Collapse
|
21
|
Qutub AA, Hunt CA. Glucose transport to the brain: a systems model. ACTA ACUST UNITED AC 2005; 49:595-617. [PMID: 16269321 DOI: 10.1016/j.brainresrev.2005.03.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2004] [Revised: 03/02/2005] [Accepted: 03/09/2005] [Indexed: 02/07/2023]
Abstract
Glucose transport to the brain involves sophisticated interactions of solutes, transporters, enzymes, and cell signaling processes, within an intricate spatial architecture. The dynamics of the transport are influenced by the adaptive nature of the blood-brain barrier (BBB), the semi-impermeable membranes of brain capillaries. As both the gate and the gatekeeper between blood-borne nutrients and brain tissue, the BBB helps govern brain homeostasis. Glucose in the blood must cross the BBB's luminal and abluminal membranes to reach neural tissue. A robust representation of the glucose transport mechanism can highlight a target for brain therapeutic intervention, help characterize mechanisms behind several disease phenotypes, or suggest a new delivery route for drugs. The challenge for researchers is understanding the relationships between influential physiological variables in vivo, and using that knowledge to predict how alterations or interventions affect glucose transport. This paper reviews factors influencing glucose transport and approaches to representing blood-to-brain glucose transport including in vitro, in vivo, and kinetic models. Applications for different models are highlighted, while their limitations in answering arising questions about the human in vivo BBB lead to a discussion of an alternate approach. A developing complex systems simulation is introduced, initiating a single platform to represent the dynamics of glucose transport across the adapting human blood-brain barrier.
Collapse
Affiliation(s)
- Amina A Qutub
- Joint Graduate Group in Bioengineering, University of California, Berkeley and San Francisco, USA.
| | | |
Collapse
|
22
|
Abstract
The memory-improving action of glucose has now been studied for almost 20 years and the study of this phenomenon has led to a number of important developments in the understanding of memory, brain physiology and pathological consequences of impaired glucose tolerance. Glucose improvement of memory appears to involve two optimal doses in animals (100 mg/kg and 2 g/kg) that may correspond to two physiological mechanisms underlying glucose effects on memory. In humans, there have been few dose-response studies so the existence of more than one effective dose in humans is uncertain. Many tasks are facilitated by glucose in humans but tasks that are difficult to master or involve divided attention are improved more readily that easier tasks. There are a number of hypotheses about the physiological bases of the memory-improving action of glucose. Peripheral glucose injections could alleviate localized deficits in extracellular glucose in the hippocampus. These localized deficits may be due to changes in glucose transporters in that structure. Because certain neurotransmitters such as acetylcholine are directly dependent on the glucose supply for their synthesis, glucose is thought to facilitate neurotransmitter synthesis under certain circumstances. However, these hypotheses cannot account for the specificity of the dose-response effect of glucose. A number of peripheral mechanisms have been proposed, including the possibility that glucose-sensitive neurons in the brain or in the periphery may serve as glucose sensors and eventually produce neural changes that would facilitate memory processing. These latter results could be of importance because the mechanisms they suggest appear to be dose-dependent, a crucial characteristic to explain the dose-dependent effects of glucose. There may be an advantage to develop hypotheses that include both peripheral and central actions of glucose. There is evidence that impaired glucose regulation is associated with impaired cognition, particularly episodic memory. This impairment is minimal in young people but increases in older people (65 years and over) where it may compound other aging processes leading to reduced brain function. A small number of studies showed that glucose improvement of memory is associated with poor glucose regulation although this may not be the case for diabetic patients. Results of a few studies also suggest that drug treatments that improve glucose regulation also produce cognitive improvement in diabetic patients.
Collapse
Affiliation(s)
- Claude Messier
- School of Psychology, University of Ottawa, 145 Jean-Jacques Lussier Room 352, Ottawa, Ontario, Canada K1N 6N5.
| |
Collapse
|
23
|
Puchowicz MA, Xu K, Magness D, Miller C, Lust WD, Kern TS, LaManna JC. Comparison of glucose influx and blood flow in retina and brain of diabetic rats. J Cereb Blood Flow Metab 2004; 24:449-57. [PMID: 15087714 DOI: 10.1097/00004647-200404000-00010] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Diabetes is associated with extensive microvascular pathology and decreased expression of the glucose transporter (GLUT-1) in retina, but not brain. To explore the basis of these differences, the authors simultaneously measured glucose influx (micromol x g(-1) x min(-1)) and blood flow (mL x g(-1) x min(-1)) in retina and brain cortex of nondiabetic control rats (normoglycemic and acute-hyperglycemic) and in rats with streptozotocin-induced diabetes (with or without aminoguanidine (AMG) treatment) using a single-pass, dual-label indicator method. In addition, tissue glucose and adenosine triphosphate (nmol/mg dry weight) levels were measured. Glucose influx in retina exceeded that of cortex by about threefold for both the nondiabetic and diabetic groups. In contrast, blood flow in retina was significantly lower than in cortex by about threefold for each group. Retinal and cortical glucose influx in the diabetic rats was lower than in the nondiabetic acute-hyperglycemic group, but not in the normoglycemic group. Blood flow in these tissues remained relatively unchanged with glycemic conditions. The glucose levels in the diabetic retina (aminoguanidine untreated and aminoguanidine treated) were fourfold to sixfold greater than the nondiabetic retina. The cortical glucose levels remained unchanged in all groups. These data suggest that the accumulation of glucose in the diabetic retina cannot be explained by increased endothelial-glucose uptake or increased vascular membrane permeability.
Collapse
Affiliation(s)
- Michelle A Puchowicz
- Department of Anatomy, Case Western Reserve University, School of Medicine, Cleveland, OH 44106-4938, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
The brain contains a small but significant amount of glycogen, which has long been considered to play an insignificant role in the brain. In this study, brain glycogen metabolism was measured using (13)C NMR spectroscopy at 9.4 T. Brain glycogen metabolism was modulated by hyperinsulinemia resulting in a net accumulation. The role of glycogen in maintaining brain function is unknown; one possibility is that it may serve as an endogenous glucose reservoir to protect the brain against severe hypoglycemia. To address this possibility, rats were subjected to insulin-induced moderate hypoglycemia and when the level of brain glucose approached zero, brain glycogen content began to decrease gradually, demonstrating utilization of this glucose reservoir. The brain glycogen signal never became undetectable, however, even during 2 hr of hypoglycemia. When plasma and brain glucose concentrations were restored, glycogen increased and the concentration exceeded the pre-hypoglycemic level by several-fold. The data suggest that brain glycogen can provide fuel for extended periods of time when glucose supply is inadequate. Furthermore, brain glycogen can rebound (super-compensate) after a single episode of hypoglycemia. We postulate that brain glycogen serves as an energy store during hypoglycemia and that it may participate in the creation of reduced physiological responses to hypoglycemia that are involved in a symptom often observed in patients with diabetes, hypoglycemia unawareness.
Collapse
Affiliation(s)
- In-Young Choi
- Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | | | | |
Collapse
|
25
|
McCall AL. Diabetes mellitus and the central nervous system. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2003; 51:415-53. [PMID: 12420366 DOI: 10.1016/s0074-7742(02)51012-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Anthony L McCall
- University of Virginia School of Medicine, Division of Endocrinology & Metabolism, Department of Internal Medicine, UVA Health System, Charlottesville, Virginia 22908, USA
| |
Collapse
|
26
|
Márián T, Balkay L, Fekete I, Lengyel Z, Veress G, Esik O, Trón L, Krasznai Z. Hypoglycemia activates compensatory mechanism of glucose metabolism of brain. ACTA BIOLOGICA HUNGARICA 2001; 52:35-45. [PMID: 11396840 DOI: 10.1556/abiol.52.2001.1.5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The effect of plasma glucose concentration on the cerebral uptake of [18F]-fluorodeoxy-D-glucose (FDG) was studied in a broad concentration range in a rabbit brain model using dynamic FDG PET measurements. Hypoglycemic and hyperglycemic conditions were maintained by manipulating plasma glucose applying i.v. glucose or insulin load. FDG utilization (K) and cerebral glucose metabolic rate (CGMR) were evaluated in a plasma glucose concentration range between 0.5 mM and 26 mM from the kinetic constant k1, k2, k3 obtained by the Sokoloff model of FDG accumulation. A decreasing set of standard FDG uptake values found with increasing blood glucose concentration was explained by competition between the plasma glucose and the radiopharmacon FDG. A similar trend was observed for the forward kinetic constants k1, and k3 in the entire concentration range studied. The same decreasing tendency of k2 was of a smaller magnitude and was reverted at the lowest glucose concentrations where a pronounced decrease of this backward transport rate constant was detected. Our kinetic data indicate a modulation of the kinetics of carbohydrate metabolism by the blood glucose concentration and report on a special mechanism compensating for the low glucose supply under conditions of extremely low blood glucose level.
Collapse
Affiliation(s)
- T Márián
- Positron Emission Tomograph Centre, University of Debrecen, Medical and Health Science Center, Hungary
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Seaquist ER, Damberg GS, Tkac I, Gruetter R. The effect of insulin on in vivo cerebral glucose concentrations and rates of glucose transport/metabolism in humans. Diabetes 2001; 50:2203-9. [PMID: 11574399 DOI: 10.2337/diabetes.50.10.2203] [Citation(s) in RCA: 134] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The continuous delivery of glucose to the brain is critically important to the maintenance of normal metabolic function. However, elucidation of the hormonal regulation of in vivo cerebral glucose metabolism in humans has been limited by the lack of direct, noninvasive methods with which to measure brain glucose. In this study, we sought to directly examine the effect of insulin on glucose concentrations and rates of glucose transport/metabolism in human brain using (1)H-magnetic resonance spectroscopy at 4 Tesla. Seven subjects participated in paired hyperglycemic (16.3 +/- 0.3 mmol/l) clamp studies performed with and without insulin. Brain glucose remained constant throughout (5.3 +/- 0.3 micromol/g wet wt when serum insulin = 16 +/- 7 pmol/l vs. 5.5 +/- 0.3 micromol/g wet wt when serum insulin = 668 +/- 81 pmol/l, P = NS). Glucose concentrations in gray matter-rich occipital cortex and white matter-rich periventricular tissue were then simultaneously measured in clamps, where plasma glucose ranged from 4.4 to 24.5 mmol/l and insulin was infused at 0.5 mU. kg(-1). min(-1). The relationship between plasma and brain glucose was linear in both regions. Reversible Michaelis-Menten kinetics fit these data best, and no differences were found in the kinetic constants calculated for each region. These data support the hypothesis that the majority of cerebral glucose uptake/metabolism is an insulin-independent process in humans.
Collapse
Affiliation(s)
- E R Seaquist
- Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA.
| | | | | | | |
Collapse
|
28
|
Régina A, Morchoisne S, Borson ND, McCall AL, Drewes LR, Roux F. Factor(s) released by glucose-deprived astrocytes enhance glucose transporter expression and activity in rat brain endothelial cells. BIOCHIMICA ET BIOPHYSICA ACTA 2001; 1540:233-42. [PMID: 11583818 DOI: 10.1016/s0167-4889(01)00133-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Glucose transporter (GLUT) expression and regulation were studied in rat brain endothelial cells in primary culture (RBEC) and in immortalised RBE4 cells. Immunoblotting analysis showed a low expression of the endothelium-specific GLUT1 in RBEC and RBE4 cells compared to isolated brain capillaries. RBEC and RBE4 cells also expressed the GLUT3 isoform, whereas it was not present in isolated brain capillaries. No change in GLUT expression was observed in endothelial cells treated with astrocyte-conditioned medium. However, treatment with conditioned medium obtained from glucose-deprived astrocytes increased endothelial GLUT1 expression and glucose uptake. These results suggest that astrocytes submitted to hypoglycaemic conditions may release factor(s) that increase glucose uptake through the blood-brain barrier.
Collapse
Affiliation(s)
- A Régina
- INSERM U26, Hôpital Fernand Widal, Paris, France
| | | | | | | | | | | |
Collapse
|
29
|
Lubec B, Chiappe-Gutierrez M, Hoeger H, Kitzmueller E, Lubec G. Glucose transporters, hexokinase, and phosphofructokinase in brain of rats with perinatal asphyxia. Pediatr Res 2000; 47:84-8. [PMID: 10625087 DOI: 10.1203/00006450-200001000-00016] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Transport by glucose transporters from blood to the brain during hypoxic-ischemic conditions is well studied. However, the recent availability of a clinically related animal model of perinatal asphyxia and the fact that no concomitant determination of glucose transporters, parameters for glucose utilization, brain glucose, and cerebral blood flow (CBF) have been reported and the early phase of perinatal asphyxia has never been studied led us to perform the following study. Cesarean section was performed on full-term pregnant rats. The obtained pups within patent uterus horns were placed into a water bath at 37 degrees C from which they were subsequently removed after 5-20 min of graded asphyxia. Brain pH, brain tissue glucose, CBF, mRNA and activity of hexokinase and phosphofructokinase, and mRNA and protein of the glucose transporters GLUTI and GLUT3 were determined. Brain pH decreased and brain tissue glucose and CBF increased with the length of the asphyctic period; hexokinase and phosphofructokinase mRNA and activity were unchanged during the observation period. The mRNA and protein of both glucose transporters were comparable between normoxic and asphyctic groups. We show that glucose transport and utilization are unchanged in the early phase of perinatal asphyxia at a time point when CBF and brain glucose are already significantly increased and severe acidosis is present.
Collapse
Affiliation(s)
- B Lubec
- Department of Pediatrics, University of Vienna, Austria
| | | | | | | | | |
Collapse
|
30
|
Abstract
Neural tissue is entirely dependent on glucose for normal metabolic activity. Since glucose stores in the brain and retina are negligible compared to glucose demand, metabolism in these tissues is dependent upon adequate glucose delivery from the systemic circulation. In the brain, the critical interface for glucose transport is at the brain capillary endothelial cells which comprise the blood-brain barrier (BBB). In the retina, transport occurs across the retinal capillary endothelial cells of the inner blood-retinal barrier (BRB) and the retinal pigment epithelium of the outer BRB. Because glucose transport across these barriers is mediated exclusively by the sodium-independent glucose transporter GLUT1, changes in endothelial glucose transport and GLUT1 abundance in the barriers of the brain and retina may have profound consequences on glucose delivery to these tissues and major implications in the development of two major diabetic complications, namely insulin-induced hypoglycemia and diabetic retinopathy. This review discusses the regulation of brain and retinal glucose transport and glucose transporter expression and considers the role of changes in glucose transporter expression in the development of two of the most devastating complications of long-standing diabetes mellitus and its management.
Collapse
Affiliation(s)
- A K Kumagai
- Department of Internal Medicine, Michigan Diabetes Research and Training Center, University of Michigan Medical School, Ann Arbor, MI 48109-0678, USA.
| |
Collapse
|
31
|
Hoxworth JM, Xu K, Zhou Y, Lust WD, LaManna JC. Cerebral metabolic profile, selective neuron loss, and survival of acute and chronic hyperglycemic rats following cardiac arrest and resuscitation. Brain Res 1999; 821:467-79. [PMID: 10064834 DOI: 10.1016/s0006-8993(98)01332-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cortical metabolites and regional cerebral intracellular pH (pHi) were measured in normoglycemic (NM), acute hyperglycemic (AH), and chronic hyperglycemic (CH, 2 week duration, streptozotocin-induced) Wistar rat brains during cardiac arrest and resuscitation. During total ischemia in AH and CH rats (plasma glucose approximately 30 mM), cortical ATP, PCr, glucose, and glycogen all fell significantly as expected. Lactate levels increased dramatically in association with a concomitant intracellular acidosis. Although lactate reached higher concentrations in AH and CH than NM, pHi was significantly lower only in the AH group. With 5 min of reperfusion, all groups recovered to near baseline in all variables, though lactate remained elevated. In a separate aspect of the study, animals from each experimental group were allowed to recover for 4 days following resuscitation, with outcome being gauged by mortality rate and hippocampal CA1 neuron counts. NM survival rate was significantly better than AH and CH. In particular, no CH rats survived for 4 days despite rapid initial recovery. After 4 days, the AH group had suffered significantly greater CA1 neuron loss than the NM rats. In summary, our research identified differences in intra-ischemic acid-base status in the two hyperglycemic groups, suggesting that chronic hyperglycemia may alter the brain's buffering capacity. These observations may account for differences between acutely and chronically hyperglycemic subjects regarding outcome, and they suggest that factors other than hydrogen ion production during ischemia are responsible for modulating outcome.
Collapse
Affiliation(s)
- J M Hoxworth
- Department of Neurology, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106-4938, USA
| | | | | | | | | |
Collapse
|
32
|
Simpson IA, Appel NM, Hokari M, Oki J, Holman GD, Maher F, Koehler-Stec EM, Vannucci SJ, Smith QR. Blood-brain barrier glucose transporter: effects of hypo- and hyperglycemia revisited. J Neurochem 1999; 72:238-47. [PMID: 9886075 DOI: 10.1046/j.1471-4159.1999.0720238.x] [Citation(s) in RCA: 206] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The transport of glucose across the blood-brain barrier (BBB) is mediated by the high molecular mass (55-kDa) isoform of the GLUT1 glucose transporter protein. In this study we have utilized the tritiated, impermeant photolabel 2-N-[4-(1 -azi-2,2,2-trifluoroethyl)[2-3H]propyl]-1,3-bis(D-mannose-4-ylo xy)-2-propylamine to develop a technique to specifically measure the concentration of GLUT1 glucose transporters on the luminal surface of the endothelial cells of the BBB. We have combined this methodology with measurements of BBB glucose transport and immunoblot analysis of isolated brain microvessels for labeled luminal GLUT1 and total GLUT1 to reevaluate the effects of chronic hypoglycemia and diabetic hyperglycemia on transendothelial glucose transport in the rat. Hypoglycemia was induced with continuous-release insulin pellets (6 U/day) for a 12- to 14-day duration; diabetes was induced by streptozotocin (65 mg/kg i.p.) for a 14- to 21-day duration. Hypoglycemia resulted in 25-45% increases in regional BBB permeability-surface area (PA) values for D-[14C]glucose uptake, when measured at identical glucose concentration using the in situ brain perfusion technique. Similarly, there was a 23+/-4% increase in total GLUT1/mg of microvessel protein and a 52+/-13% increase in luminal GLUT1 in hypoglycemic animals, suggesting that both increased GLUT1 synthesis and a redistribution to favor luminal transporters account for the enhanced uptake. A corresponding (twofold) increase in cortical GLUT1 mRNA was observed by in situ hybridization. In contrast, no significant changes were observed in regional brain glucose uptake PA, total microvessel 55-kDa GLUT1, or luminal GLUT1 concentrations in hyperglycemic rats. There was, however, a 30-40% increase in total cortical GLUT1 mRNA expression, with a 96% increase in the microvessels. Neither condition altered the levels of GLUT3 mRNA or protein expression. These results show that hypoglycemia, but not hyperglycemia, alters glucose transport activity at the BBB and that these changes in transport activity result from both an overall increase in total BBB GLUT1 and an increased transporter concentration at the luminal surface.
Collapse
Affiliation(s)
- I A Simpson
- Experimental Diabetes, Metabolism, and Nutrition Section, NIDDK, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Blomqvist G, Grill V, Ingvar M, Widén L, Stone-Elander S. The effect of hyperglycaemia on regional cerebral glucose oxidation in humans studied with [1-11C]-D-glucose. ACTA PHYSIOLOGICA SCANDINAVICA 1998; 163:403-15. [PMID: 9789584 DOI: 10.1046/j.1365-201x.1998.t01-1-00360.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The effect of hyperglycaemia on regional cerebral glucose utilization was studied in five healthy males fasted over-night using positron emission tomography. Selectively labelled glucose, [1-11C]-D-glucose, was used as a tracer. After correction for the small loss of [11C]CO2 from the tissue, this tracer measures the rate of glucose oxidation rather than the total rate of glucose metabolism. Each subject was investigated twice: during normoglycaemia (plasma glucose 5.3 +/- 0.3 mumol mL-1) and at the end of a 2-h period of hyperglycaemia (plasma glucose 13.8 +/- 0.7 mumol mL-1). Assuming unchanged rate constant for loss of labelled CO2 at normo- and hyperglycaemia the oxidative metabolic rate of glucose was found to be slightly larger at combined hyperglycaemia and hypersulinemia (0.30 +/- 0.01 mmol mL-1 min-1) than at normal glucose and insulin levels (0.25 +/- 0.01 mmol mL-1 min-1). This suggests that the process of glucose phosphorylation might not be fully saturated in the human brain or, alternatively, that the glycogen deposition increases during short-term hyperglycaemia. The relative increase of oxidative metabolic rate was considerably larger (approximately 50%) in white matter than in the brain as a whole (20%). The brain glucose content was found to increase non-linearly with increasing plasma glucose. Together with data from previous studies these results suggest that the free glucose in the human brain is close to zero when the plasma glucose is below 2 mumol mL-1.
Collapse
Affiliation(s)
- G Blomqvist
- INSERM U334, Service Hospitalier Frédéric Joliot, Orsay, France
| | | | | | | | | |
Collapse
|
34
|
Cobbs CS, Chen J, Greenberg DA, Graham SH. Vascular endothelial growth factor expression in transient focal cerebral ischemia in the rat. Neurosci Lett 1998; 249:79-82. [PMID: 9682821 DOI: 10.1016/s0304-3940(98)00377-2] [Citation(s) in RCA: 100] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Vascular endothelial growth factor (VEGF) has been implicated in hypoxia-induced angiogenesis in tumors and ischemia. We examined VEGF mRNA and protein expression after occlusion of the middle cerebral artery (MCA) in rats. VEGF mRNA expression studied by in situ hybridization was increased in the ischemic border zone 24 h after 30, 60 or 120 min of focal cerebral ischemia. VEGF protein expression measured by Western blots was also increased in this region 24 and 48 h after ischemia, and VEGF immunocytochemistry localized this increased expression to astroglia. Thus, VEGF is induced after focal cerebral ischemia and could have a role in pathophysiology and recovery in the ischemic border zone.
Collapse
Affiliation(s)
- C S Cobbs
- Department of Neurosurgery, University of Alabama, Birmingham, USA
| | | | | | | |
Collapse
|
35
|
Chipkin SR, van Bueren A, Bercel E, Garrison CR, McCall AL. Effects of dexamethasone in vivo and in vitro on hexose transport in brain microvasculature. Neurochem Res 1998; 23:645-52. [PMID: 9566602 DOI: 10.1023/a:1022434721114] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Glucocorticoids induce hyperinsulinemia, hyperglycemia, and depress glucose transport by aortic endothelium. High glucocorticoid doses are used for many diseases, but with unknown effects on brain glucose transport or metabolism. This study tested the hypothesis that glucocorticoids affect glucose transport or metabolism by brain microvascular endothelium. Male rats received dexamethasone (DEX) s.c. with sucrose feeding for up to seven days. Cerebral microvessels from rats treated with DEX/sucrose demonstrated increased GLUT1 and brain glucose extraction compared to controls. Glucose transport in vivo correlated with hyperinsulinemia. Pre-treatment with low doses of streptozotocin blunted hyperinsulinemia and prevented increased glucose extraction induced by DEX. In contrast, isolated brain microvessels exposed to DEX in vitro demonstrated suppression of 2-deoxyglucose uptake and glucose oxidation. We conclude that DEX/sucrose treatment in vivo increases blood-brain glucose transport in a manner that requires the effects of chronic hyperinsulinemia. These effects override any direct inhibitory effects of either hyperglycemia or DEX.
Collapse
Affiliation(s)
- S R Chipkin
- Department of Medicine, Boston University School of Medicine, MA 02118, USA
| | | | | | | | | |
Collapse
|
36
|
|
37
|
Abstract
Ischemic brain injury is the third-leading cause of death among Americans and the leading cause of serious disability. Based on studies of animal models, a substantial amount of experimental evidence shows that hyperglycemia at the onset of brain ischemia worsens postischemic neurologic outcome. Consistent with these observations, hyperglycemia also is associated with a worsening of postischemic brain injury in humans. In humans, however, data are often difficult to interpret because of problems in determining the timing of hyperglycemia relative to a critical ischemic event and in elucidating the effect of coexisting pathophysiologic processes (for example, a stress response) on outcome. Glucose modulation of neurologic injury is observed when ischemia is either global (for example, that accompanying cardiac arrest or severe systemic hypotension) or focal (for example, that accompanying thrombotic or embolic stroke). Toxicity is probably the result of an intracellular lactic acidosis. Specifically, the associated hydrogen ions are injurious to neurons and glia. On the basis of these factors, we recommend diligent monitoring of blood glucose concentrations in patients who are at increased risk for new-onset, ongoing, or recurring cerebral ischemia. In such patients, the use of fluid infusions, corticosteroid drugs, and insulin, as well as stress management, should be tailored to treat preexisting hyperglycemia and prevent new-onset hyperglycemia. Maintenance of normoglycemia is recommended. When one attempts to treat preexisting hyperglycemia, care should be taken to avoid rapid fluid shifts, electrolyte abnormalities, and hypoglycemia, all of which can be detrimental to the brain.
Collapse
Affiliation(s)
- C T Wass
- Department of Anesthesiology, Mayo Clinic Rochester, MN 55905, USA
| | | |
Collapse
|
38
|
Gruetter R, Garwood M, Uğurbil K, Seaquist ER. Observation of resolved glucose signals in 1H NMR spectra of the human brain at 4 Tesla. Magn Reson Med 1996; 36:1-6. [PMID: 8795012 DOI: 10.1002/mrm.1910360102] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Measurement of the resonances of glucose between 3.2 and 3.9 ppm in 1H NMR spectra from the human brain is difficult due to spectral overlap with peaks from more concentrated metabolites. The H1 resonance of alpha-D-glucose at 5.23 ppm is resolved from other metabolite peaks, but potentially overlaps with the intense water signal at 4.72 ppm. This paper demonstrates that the increased resolution at 4 Tesla permits to suppress the water signal sufficiently to reliably detect glucose directly at 5.23 ppm by 1H MRS and the estimated peak intensity is consistent with previous 13C NMR quantification.
Collapse
Affiliation(s)
- R Gruetter
- Department of Medicine, University of Minnesota, Minneapolis 55455, USA
| | | | | | | |
Collapse
|
39
|
Gruetter R, Novotny EJ, Boulware SD, Rothman DL, Shulman RG. 1H NMR studies of glucose transport in the human brain. J Cereb Blood Flow Metab 1996; 16:427-38. [PMID: 8621747 DOI: 10.1097/00004647-199605000-00009] [Citation(s) in RCA: 82] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The difference between 1H nuclear magnetic resonance (NMR) spectra obtained from the human brain during euglycemia and during hyperglycemia is depicted as well-resolved glucose peaks. The time course of these brain glucose changes during a rapid increase in plasma glucose was measured in four healthy subjects, aged 18-22 years, in five studies. Results demonstrated a significant lag in the rise of glucose with respect to plasma glucose. The fit of the integrated symmetric Michaelis-Menten model to the time course of relative glucose signals yielded an estimated plasma glucose concentration for half maximal transport, Kt, of 4.8 +/- 2.4 mM (mean +/- SD), a maximal transport rate, Tmax, of 0.80 +/- 0.45 micromol g-1 min-1, and a cerebral metabolic glucose consumption rate (CMR)glc of 0.32 +/- 0.16 micromol g-1 min-1. Assuming cerebral glucose concentration to be 1.0 micromol/g at euglycemia as measured by 13CMR, the fit of the same model to the time course of brain glucose concentrations resulted in Kt = 3.9 +/- 0.82 mM, Tmax = 1.16 +/- 0.29 micromol g-1 min-1, and CMRglc = 0.35 +/- 0.10 micromol g-1 min-1. In both cases, the resulting time course equaled that predicted from the determination of the steady-state glucose concentration by 13C NMR spectroscopy within the experimental scatter. The agreement between the two methods of determining transport kinetics suggests that glucose is distributed throughout the entire aqueous phase of the human brain, implying substantial intracellular concentration.
Collapse
Affiliation(s)
- R Gruetter
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
| | | | | | | | | |
Collapse
|
40
|
Messier C, Gagnon M. Glucose regulation and cognitive functions: relation to Alzheimer's disease and diabetes. Behav Brain Res 1996; 75:1-11. [PMID: 8800646 DOI: 10.1016/0166-4328(95)00153-0] [Citation(s) in RCA: 164] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Glucose has been found to improve memory in animals and humans. Animal research has revealed that glucose may improve memory through a facilitation of acetylcholine (ACh) synthesis and release in the brain. This glucose-related memory improvement has prompted research in elderly humans. These studies have shown that the memory-improving action of glucose depends on each individuals' blood glucose regulation. Based on these data, researchers have evaluated the effect of glucose on memory in patients with Alzheimer's disease (AD). Results demonstrated that glucose could improve memory in a subset of patients that had abnormalities in their blood glucose regulation. Interestingly, these alterations in blood glucose regulation were believed to depend on the severity of the disease process. Another line of investigation has focused on alterations in brain glucose metabolism. Both animal models and studies with Type II diabetic elderly patients have shown that altered glucose regulation impairs learning and memory processes. It is possible that in AD patients, hyperglycemia exerts a deleterious effect by potentiating the neuronal death produced by other pathological processes taking place such as amyloid deposition. Based on these data, it appears important to find the prevalence of altered glucoregulation at various stages of AD. Secondly, it may be of interest to determine prospectively whether altered glucoregulation is linked to a faster progression of the disease. Finally, if such a relationship is observed, the next logical step would be to determine whether AD patients could benefit from treatments aimed at normalizing blood glucose regulation and improving insulin sensitivity.
Collapse
Affiliation(s)
- C Messier
- School of Psychology, University of Ottawa, Ontario, Canada.
| | | |
Collapse
|
41
|
|
42
|
Powers WJ, Dagogo-Jack S, Markham J, Larson KB, Dence CS. Cerebral transport and metabolism of 1-11C-D-glucose during stepped hypoglycemia. Ann Neurol 1995; 38:599-609. [PMID: 7574456 DOI: 10.1002/ana.410380408] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Attempts to measure blood-to-brain glucose transport and cerebral glucose metabolism with 11C-glucose have been hampered by methods that require jugular venous sampling or do not adequately account for the efflux of labeled metabolites from the brain. We performed eight positron emission tomography studies with 1-11C-D-glucose in macaques at arterial plasma glucose concentrations of 8.43 to 1.51 mumol ml-1 (152-27 mg dl-1) using a model that includes a fourth rate constant to account for regional egress of all 11C-metabolites. Values for blood-to-brain glucose influx, cerebral glucose metabolism, and brain free glucose concentration agreed closely with values obtained in mammals by other investigators. Values for net extraction fraction corresponded closely to simultaneously measured arteriovenous values. We demonstrated that utilization of a model that includes a fourth rate constant to account for regional egress of all 11C-metabolites with positron emission tomography and 1-11C-D-glucose provides accurate measurements of blood-to-brain glucose transport and cerebral glucose metabolism in vivo without need for jugular venous sampling, even under conditions of severe hypoglycemia.
Collapse
Affiliation(s)
- W J Powers
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
43
|
Whitesell RR, Ward M, McCall AL, Granner DK, May JM. Coupled glucose transport and metabolism in cultured neuronal cells: determination of the rate-limiting step. J Cereb Blood Flow Metab 1995; 15:814-26. [PMID: 7673374 DOI: 10.1038/jcbfm.1995.102] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
In brain and nerves the phosphorylation of glucose, rather than its transport, is generally considered the major rate-limiting step in metabolism. Since little is known regarding the kinetic coupling between these processes in neuronal tissues, we investigated the transport and phosphorylation of [2-3H]glucose in two neuronal cell models: a stable neuroblastoma cell line (NCB20), and a primary culture of isolated rat dorsal root ganglia cells. When transport and phosphorylation were measured in series, phosphorylation was the limiting step, because intracellular glucose concentrations were the same as those outside of cells, and because the apparent Km for glucose utilization was lower than expected for the transport step. However, the apparent Km was still severalfold higher than the Km of hexokinase I. When [2-3H]glucose efflux and phosphorylation were measured from the same intracellular glucose pool in a parallel assay, rates of glucose efflux were three- to-fivefold greater than rates of phosphorylation. With the parallel assay, we observed that activation of glucose utilization by the sodium channel blocker veratridine caused a selective increase in glucose phosphorylation and was without effect on glucose transport. In contrast to results with glucose, both cell types accumulated 2-deoxy-D-[14C]glucose to concentrations severalfold greater than extracellular concentrations. We conclude from these studies that glucose utilization in neuronal cells is phosphorylation-limited, and that the coupling between transport and phosphorylation depends on the type of hexose used.
Collapse
Affiliation(s)
- R R Whitesell
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232-2230, USA
| | | | | | | | | |
Collapse
|
44
|
Rahner-Welsch S, Vogel J, Kuschinsky W. Regional congruence and divergence of glucose transporters (GLUT1) and capillaries in rat brains. J Cereb Blood Flow Metab 1995; 15:681-6. [PMID: 7790418 DOI: 10.1038/jcbfm.1995.84] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The association of glucose transporters (GLUT1) and brain capillaries was tested in different brain structures of rats by a direct comparison of the topologies of capillaries and GLUT1 in identical brain sections. Antibody staining of capillaries (fibronectin) and GLUT1 were made visible by fluorescence microscopy. The results showed differences between brain structures containing a tight and a leaky blood-brain barrier. All capillaries of brain structures with a tight blood-brain barrier showed congruent staining of GLUT1 and capillary morphology. The circumventricular organs that are known to have leaky barrier capillaries were stained by fibronectin antibodies but not by GLUT1 antibodies. Ependymal cells showed moderate staining by GLUT1 antibodies both in areas with tight and leaky barriers. The subcommissural organ appeared to be unique showing neither capillary nor GLUT1 stain. It is concluded that glucose transporters (GLUT1) exist in all brain capillaries of blood-brain barrier structures, whereas they are absent in leaky barrier structures. Moderate amounts of glucose transporter (GLUT1) can also be detected in ependymal cells.
Collapse
|
45
|
Hasselbalch SG, Knudsen GM, Jakobsen J, Hageman LP, Holm S, Paulson OB. Blood-brain barrier permeability of glucose and ketone bodies during short-term starvation in humans. THE AMERICAN JOURNAL OF PHYSIOLOGY 1995; 268:E1161-6. [PMID: 7611392 DOI: 10.1152/ajpendo.1995.268.6.e1161] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The blood-brain barrier (BBB) permeability for glucose and beta-hydroxybutyrate (beta-OHB) was studied by the intravenous double-indicator method in nine healthy subjects before and after 3.5 days of starvation. In fasting, mean arterial plasma glucose decreased and arterial concentration of beta-OHB increased, whereas cerebral blood flow remained unchanged. The permeability-surface area product for BBB glucose transport from blood to brain (PS1) increased by 55 +/- 31%, whereas no significant change in the permeability from brain back to blood (PS2) was found. PS1 for beta-OHB remained constant during starvation. The expected increase in PS1 due to the lower plasma glucose concentration was calculated to be 22% using previous estimates of maximal transport velocity and Michaelis-Menten affinity constant for glucose transport. The determined increase was thus 33% higher than the expected increase and can only be partially explained by the decrease in plasma glucose. It is concluded that a modest upregulation of glucose transport across the BBB takes place after starvation. Brain transport of beta-OHB did not decrease as expected from the largely increased beta-OHB arterial level. This might be interpreted as an increase in brain transport of beta-OHB, which could be caused by induction mechanisms, but the large nonsaturable component of beta-OHB transport makes such a conclusion difficult. However, beta-OHB blood concentration and beta-OHB influx into the brain increased by > 10 times. This implies that the influx of ketone bodies into the brain is largely determined by the amount of ketones present in the blood, and any condition in which ketonemia occurs will lead to an increased ketone influx.
Collapse
Affiliation(s)
- S G Hasselbalch
- Department of Neurology, University of Hospital, Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
46
|
Xia Y, Warshaw JB, Haddad GG. Chronic hypoxia causes opposite effects on glucose transporter 1 mRNA in mature versus immature rat brain. Brain Res 1995; 675:224-30. [PMID: 7796133 DOI: 10.1016/0006-8993(95)00079-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
We have shown previously that chronic hypoxia can regulate the expression of membrane proteins. Since there are virtually no glucose stores in the brain and glucose transport can be rate-limiting during stress, the role of glucose transporters becomes crucial for cell survival under stress. In the present study, we asked whether mRNA levels for glucose transporter 1 (GT1), which is expressed in a variety of cells in the brain, especially in the microvessels for glucose transport from blood vessels to brain, change in response to chronic hypoxia. Because major developmental changes occur in the rat CNS in-utero and in the first few weeks postnatally, we studied brain GT1 mRNA using Northern blot analysis at different ages after exposure of fetuses (from embryonic day 10 to birth), developing rats (from birth to 30 day old) or adult rats (from 90 to 120 day old) to hypoxia (Fractional inspired O2 9%). Our data show that (i) GT1 mRNA level was much lower in the newborn than in the adult and increased with age; (ii) chronic hypoxia caused a decrease of approximately 65% in GT1 mRNA in adult brain but induced an increase in fetal (more than 50%) and developing (approximately 80%) rats and (iii) the response of housekeeping gene (glyceraldehyde 3-phosphate dehydrogenase) was not similar to that of GT1, suggesting that the changes of GT1 mRNA are specific to glucose transporter.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- Y Xia
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | |
Collapse
|
47
|
Archer DP, Ravussin PA. [Role of blood-brain barrier in cerebral homeostasis]. ANNALES FRANCAISES D'ANESTHESIE ET DE REANIMATION 1994; 13:57-61. [PMID: 7916553 DOI: 10.1016/s0750-7658(94)80187-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
By a variety of mechanisms, the cerebral endothelium isolates the extracellular fluid space in the central nervous system from the plasma. The combination of physical and enzymatic mechanisms which prevent macromolecules, polar solutes, neurotransmitters, peptides, and electrolytes from passively entering the brain has been termed the blood-brain barrier (BBB). Specific mechanisms provide facilitated transport across the BBB and active secretion of extracellular fluid and CSF maintain homeostasis for nutrients and for cation and H+ respectively. Consequently, interstitial fluid volume in the CNS does not increase when the total extracellular fluid volume is increased. Total tissue volume is sensitive to osmotic forces, while oncotic forces are relatively unimportant. Most anaesthetic drugs are sufficiently lipid soluble that they enter the CNS easily by passive diffusion. Differences in the rates of CNS penetration between drugs can be predicted from their lipid solubility. Anaesthetic drugs have little effect on BBB permeability and their effects on brain oedema formation derive principally from their haemodynamics effects.
Collapse
Affiliation(s)
- D P Archer
- Department of Anaesthesia, Foothills Hospital, Calgary, Alberta, Canada
| | | |
Collapse
|
48
|
Kainulainen H, Schürmann A, Vilja P, Joost HG. In-vivo glucose uptake and glucose transporter proteins GLUT1 and GLUT3 in brain tissue from streptozotocin-diabetic rats. ACTA PHYSIOLOGICA SCANDINAVICA 1993; 149:221-5. [PMID: 8266811 DOI: 10.1111/j.1748-1716.1993.tb09615.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The effects of streptozotocin-induced diabetes (13 weeks) on the in-vivo glucose uptake and on the protein levels of glucose transporters in rat brain were studied and compared with those in cardiac muscle. Diabetes reduced the uptake of 2-[3H]deoxyglucose into lobus frontalis by 70%. However, uptake rates corrected for the 4-fold increase in serum glucose (glucose metabolic index, GMI) were essentially unaltered. The levels of glucose transporter proteins GLUT1 and GLUT3 in crude membranes from brain as assessed by immunoblotting were unaffected by diabetes, whereas GMI and levels of glucose transporters GLUT1 and GLUT4 in heart were reduced by 80 and 65%, respectively. Thus, glucose uptake and levels of glucose transporters in brain, unlike that in insulin sensitive tissues, are normal in long-term hypo-insulinaemia.
Collapse
Affiliation(s)
- H Kainulainen
- Institute of Pharmacology and Toxicology, RWTH Aachen, Germany
| | | | | | | |
Collapse
|
49
|
LaManna JC, Harrington JF, Vendel LM, Abi-Saleh K, Lust WD, Harik SI. Regional blood-brain lactate influx. Brain Res 1993; 614:164-70. [PMID: 8348311 DOI: 10.1016/0006-8993(93)91030-v] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Regional blood-to-brain lactate transport was studied in chloral hydrate anesthetized rats using the single pass, dual-label, indicator fractionation, right atrial injection method. Lactate influx was resolved into two components, a saturable, stereospecific (to the L-enantiomer) component and a non-saturable, non-stereospecific diffusional component. The saturable component was found to have a low efficiency and moderate capacity with transport affinity coefficients between 6 and 14 mM and transport maxima of 23-40 mumol/100 g/min in the various brain regions. Lactate transport was not inhibited by probenecid. The diffusional component was determined from D-lactate influx measurements and the regional linear diffusion coefficients ranged from 0.020 to 0.036 ml/g/min. At the usual levels of plasma lactate (1-1.5 mM) these two influx components were about equal. The relative contribution of the non-stereospecific diffusional component was increased at higher plasma lactate concentrations. Lactate clearance, estimated by the total apparent permeability x surface area products was between 6 and 8 ml/100 g/min.
Collapse
Affiliation(s)
- J C LaManna
- Department of Neurology, University Hospitals of Cleveland, OH
| | | | | | | | | | | |
Collapse
|