1
|
Liang R, Hou X, Zhou D, Zhu L, Teng L, Song W, Tang Q. Exercise preconditioning mitigates Ischemia-Reperfusion injury in rats by enhancing mitochondrial respiration. Neuroscience 2024; 562:64-74. [PMID: 39461659 DOI: 10.1016/j.neuroscience.2024.10.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 10/29/2024]
Abstract
Cerebral ischemia and subsequent reperfusion damage are prevalent in clinical practice, linked to numerous neurodegenerative diseases. Cerebral ischemia deprives brain tissue of essential oxygen and nutrients, disrupting energy metabolism and causing cellular dysfunction. Although reperfusion theoretically aids recovery, it instead initiates complex injury responses such as oxidative stress, apoptosis, and inflammation, worsening brain damage. Recent research suggests that enhancing neuronal energy status by modulating energy metabolism pathways can effectively counter these effects. For instance, boosting mitochondrial function, improving energy provision, and decreasing harmful metabolites can mitigate oxidative stress and cellular injury. This study investigated the protective effects of exercise preconditioning against ischemia-reperfusion injury in rats. It was observed that exercise enhances energy levels and mitochondrial respiration by upregulating the expression of COX4 and NAMPT proteins and activating AMPK and mitochondrial complex V. This process facilitates metabolic reprogramming characterized by the promotion of oxidative phosphorylation (OXPHOS) and the pentose phosphate pathway (PPP), alongside a reduction in glycolysis. Such reprogramming reduces harmful metabolites, mitigating apoptosis and oxidative stress, and is a key factor in alleviating acute ischemic hypoxia-induced brain damage. These findings introduce a novel therapeutic approach for ischemic brain reperfusion injury, underscoring the crucial role of ATP production and metabolic regulation in neuroprotection.
Collapse
Affiliation(s)
- Runyu Liang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xinlei Hou
- Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Daguo Zhou
- Xiang'an Hospital of Xiamen University, China
| | - Luwen Zhu
- Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lili Teng
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wenjing Song
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qiang Tang
- Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China.
| |
Collapse
|
2
|
Radford-Smith D, Ng TT, Yates AG, Dunstan I, Claridge TDW, Anthony DC, Probert F. Ex-Vivo 13C NMR Spectroscopy of Rodent Brain: TNF Restricts Neuronal Utilization of Astrocyte-Derived Metabolites. J Proteome Res 2024; 23:3383-3392. [PMID: 38943617 PMCID: PMC11301676 DOI: 10.1021/acs.jproteome.4c00035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 07/01/2024]
Abstract
Tumor necrosis factor (TNF) has well-established roles in neuroinflammatory disorders, but the effect of TNF on the biochemistry of brain cells remains poorly understood. Here, we microinjected TNF into the brain to study its impact on glial and neuronal metabolism (glycolysis, pentose phosphate pathway, citric acid cycle, pyruvate dehydrogenase, and pyruvate carboxylase pathways) using 13C NMR spectroscopy on brain extracts following intravenous [1,2-13C]-glucose (to probe glia and neuron metabolism), [2-13C]-acetate (probing astrocyte-specific metabolites), or [3-13C]-lactate. An increase in [4,5-13C]-glutamine and [2,3-13C]-lactate coupled with a decrease in [4,5-13C]-glutamate was observed in the [1,2-13C]-glucose-infused animals treated with TNF. As glutamine is produced from glutamate by astrocyte-specific glutamine synthetase the increase in [4,5-13C]-glutamine reflects increased production of glutamine by astrocytes. This was confirmed by infusion with astrocyte substrate [2-13C]-acetate. As lactate is metabolized in the brain to produce glutamate, the simultaneous increase in [2,3-13C]-lactate and decrease in [4,5-13C]-glutamate suggests decreased lactate utilization, which was confirmed using [3-13C]-lactate as a metabolic precursor. These results suggest that TNF rearranges the metabolic network, disrupting the energy supply chain perturbing the glutamine-glutamate shuttle between astrocytes and the neurons. These insights pave the way for developing astrocyte-targeted therapeutic strategies aimed at modulating effects of TNF to restore metabolic homeostasis in neuroinflammatory disorders.
Collapse
Affiliation(s)
- Daniel Radford-Smith
- Department
of Chemistry, University of Oxford, Oxford OX1 3TA, U.K.
- Pharmacology
Department, University of Oxford, Oxford OX1 3QT, U.K.
| | - Tang T. Ng
- Department
of Chemistry, University of Oxford, Oxford OX1 3TA, U.K.
| | - Abi G. Yates
- Department
of Chemistry, University of Oxford, Oxford OX1 3TA, U.K.
- Pharmacology
Department, University of Oxford, Oxford OX1 3QT, U.K.
| | - Isobel Dunstan
- Pharmacology
Department, University of Oxford, Oxford OX1 3QT, U.K.
| | | | | | - Fay Probert
- Department
of Chemistry, University of Oxford, Oxford OX1 3TA, U.K.
| |
Collapse
|
3
|
Peper CJ, Kilgore MD, Jiang Y, Xiu Y, Xia W, Wang Y, Shi M, Zhou D, Dumont AS, Wang X, Liu N. Tracing the path of disruption: 13C isotope applications in traumatic brain injury-induced metabolic dysfunction. CNS Neurosci Ther 2024; 30:e14693. [PMID: 38544365 PMCID: PMC10973562 DOI: 10.1111/cns.14693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/25/2024] [Accepted: 03/12/2024] [Indexed: 05/14/2024] Open
Abstract
Cerebral metabolic dysfunction is a critical pathological hallmark observed in the aftermath of traumatic brain injury (TBI), as extensively documented in clinical investigations and experimental models. An in-depth understanding of the bioenergetic disturbances that occur following TBI promises to reveal novel therapeutic targets, paving the way for the timely development of interventions to improve patient outcomes. The 13C isotope tracing technique represents a robust methodological advance, harnessing biochemical quantification to delineate the metabolic trajectories of isotopically labeled substrates. This nuanced approach enables real-time mapping of metabolic fluxes, providing a window into the cellular energetic state and elucidating the perturbations in key metabolic circuits. By applying this sophisticated tool, researchers can dissect the complexities of bioenergetic networks within the central nervous system, offering insights into the metabolic derangements specific to TBI pathology. Embraced by both animal studies and clinical research, 13C isotope tracing has bolstered our understanding of TBI-induced metabolic dysregulation. This review synthesizes current applications of isotope tracing and its transformative potential in evaluating and addressing the metabolic sequelae of TBI.
Collapse
Affiliation(s)
- Charles J. Peper
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Mitchell D. Kilgore
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Yinghua Jiang
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Yuwen Xiu
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Winna Xia
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Yingjie Wang
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Mengxuan Shi
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Di Zhou
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Aaron S. Dumont
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Xiaoying Wang
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
- Neuroscience Program, Tulane Brain InstituteTulane UniversityNew OrleansLouisianaUSA
| | - Ning Liu
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
- Neuroscience Program, Tulane Brain InstituteTulane UniversityNew OrleansLouisianaUSA
- Tulane University Translational Sciences InstituteNew OrleansLouisianaUSA
| |
Collapse
|
4
|
Gribnau A, van Zuylen ML, Coles JP, Plummer MP, Hermanns H, Hermanides J. Cerebral Glucose Metabolism following TBI: Changes in Plasma Glucose, Glucose Transport and Alternative Pathways of Glycolysis-A Translational Narrative Review. Int J Mol Sci 2024; 25:2513. [PMID: 38473761 DOI: 10.3390/ijms25052513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/05/2024] [Accepted: 02/14/2024] [Indexed: 03/14/2024] Open
Abstract
Traumatic brain injury (TBI) is a major public health concern with significant consequences across various domains. Following the primary event, secondary injuries compound the outcome after TBI, with disrupted glucose metabolism emerging as a relevant factor. This narrative review summarises the existing literature on post-TBI alterations in glucose metabolism. After TBI, the brain undergoes dynamic changes in brain glucose transport, including alterations in glucose transporters and kinetics, and disruptions in the blood-brain barrier (BBB). In addition, cerebral glucose metabolism transitions from a phase of hyperglycolysis to hypometabolism, with upregulation of alternative pathways of glycolysis. Future research should further explore optimal, and possibly personalised, glycaemic control targets in TBI patients, with GLP-1 analogues as promising therapeutic candidates. Furthermore, a more fundamental understanding of alterations in the activation of various pathways, such as the polyol and lactate pathway, could hold the key to improving outcomes following TBI.
Collapse
Affiliation(s)
- Annerixt Gribnau
- Department of Anaesthesiology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Mark L van Zuylen
- Department of Anaesthesiology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Department of Paediatric Intensive Care, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Jonathan P Coles
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Mark P Plummer
- Intensive Care Unit, Royal Melbourne Hospital, 300 Grattan Street, Parkville, VIC 3050, Australia
| | - Henning Hermanns
- Department of Anaesthesiology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Jeroen Hermanides
- Department of Anaesthesiology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
5
|
Hackett EP, Chen J, Ingle L, Nemri SA, Barshikar S, da Cunha Pinho M, Plautz EJ, Bartnik-Olson BL, Park JM. Longitudinal assessment of mitochondrial dysfunction in acute traumatic brain injury using hyperpolarized [1- 13 C]pyruvate. Magn Reson Med 2023; 90:2432-2442. [PMID: 37427535 PMCID: PMC10543630 DOI: 10.1002/mrm.29794] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/11/2023]
Abstract
PURPOSE [13 C]Bicarbonate formation from hyperpolarized [1-13 C]pyruvate via pyruvate dehydrogenase, a key regulatory enzyme, represents the cerebral oxidation of pyruvate and the integrity of mitochondrial function. The present study is to characterize the chronology of cerebral mitochondrial metabolism during secondary injury associated with acute traumatic brain injury (TBI) by longitudinally monitoring [13 C]bicarbonate production from hyperpolarized [1-13 C]pyruvate in rodents. METHODS Male Wistar rats were randomly assigned to undergo a controlled-cortical impact (CCI, n = 31) or sham surgery (n = 22). Seventeen of the CCI and 9 of the sham rats longitudinally underwent a 1 H/13 C-integrated MR protocol that includes a bolus injection of hyperpolarized [1-13 C]pyruvate at 0 (2 h), 1, 2, 5, and 10 days post-surgery. Separate CCI and sham rats were used for histological validation and enzyme assays. RESULTS In addition to elevated lactate, we observed significantly reduced bicarbonate production in the injured site. Unlike the immediate appearance of hyperintensity on T2 -weighted MRI, the contrast of bicarbonate signals between the injured region and the contralateral brain peaked at 24 h post-injury, then fully recovered to the normal level at day 10. A subset of TBI rats demonstrated markedly increased bicarbonate in normal-appearing contralateral brain regions post-injury. CONCLUSION This study demonstrates that aberrant mitochondrial metabolism occurring in acute TBI can be monitored by detecting [13 C]bicarbonate production from hyperpolarized [1-13 C]pyruvate, suggesting that [13 C]bicarbonate is a sensitive in-vivo biomarker of the secondary injury processes.
Collapse
Affiliation(s)
- Edward P. Hackett
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas TX USA 75390
| | - Jun Chen
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas TX USA 75390
| | - Laura Ingle
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas TX USA 75390
| | - Sarah Al Nemri
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas TX USA 75390
| | - Surendra Barshikar
- Department of Physical Medicine and Rehabilitation, The University of Texas Southwestern Medical Center, Dallas TX USA 75390
| | - Marco da Cunha Pinho
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas TX USA 75390
| | - Erik J. Plautz
- Department of Neurology and Neurotherapeutics, The University of Texas Southwestern Medical Center, Dallas TX USA 75390
| | | | - Jae Mo Park
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas TX USA 75390
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas TX USA 75390
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas TX USA 75390
| |
Collapse
|
6
|
Strogulski NR, Portela LV, Polster BM, Loane DJ. Fundamental Neurochemistry Review: Microglial immunometabolism in traumatic brain injury. J Neurochem 2023; 167:129-153. [PMID: 37759406 PMCID: PMC10655864 DOI: 10.1111/jnc.15959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023]
Abstract
Traumatic brain injury (TBI) is a devastating neurological disorder caused by a physical impact to the brain that promotes diffuse damage and chronic neurodegeneration. Key mechanisms believed to support secondary brain injury include mitochondrial dysfunction and chronic neuroinflammation. Microglia and brain-infiltrating macrophages are responsible for neuroinflammatory cytokine and reactive oxygen species (ROS) production after TBI. Their production is associated with loss of homeostatic microglial functions such as immunosurveillance, phagocytosis, and immune resolution. Beyond providing energy support, mitochondrial metabolic pathways reprogram the pro- and anti-inflammatory machinery in immune cells, providing a critical immunometabolic axis capable of regulating immunologic response to noxious stimuli. In the brain, the capacity to adapt to different environmental stimuli derives, in part, from microglia's ability to recognize and respond to changes in extracellular and intracellular metabolite levels. This capacity is met by an equally plastic metabolism, capable of altering immune function. Microglial pro-inflammatory activation is associated with decreased mitochondrial respiration, whereas anti-inflammatory microglial polarization is supported by increased oxidative metabolism. These metabolic adaptations contribute to neuroimmune responses, placing mitochondria as a central regulator of post-traumatic neuroinflammation. Although it is established that profound neurometabolic changes occur following TBI, key questions related to metabolic shifts in microglia remain unresolved. These include (a) the nature of microglial mitochondrial dysfunction after TBI, (b) the hierarchical positions of different metabolic pathways such as glycolysis, pentose phosphate pathway, glutaminolysis, and lipid oxidation during secondary injury and recovery, and (c) how immunometabolism alters microglial phenotypes, culminating in chronic non-resolving neuroinflammation. In this basic neurochemistry review article, we describe the contributions of immunometabolism to TBI, detail primary evidence of mitochondrial dysfunction and metabolic impairments in microglia and macrophages, discuss how major metabolic pathways contribute to post-traumatic neuroinflammation, and set out future directions toward advancing immunometabolic phenotyping in TBI.
Collapse
Affiliation(s)
- Nathan R. Strogulski
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Luis V. Portela
- Neurotrauma and Biomarkers Laboratory, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Brian M. Polster
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - David J. Loane
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
7
|
Stovell MG, Howe DJ, Thelin EP, Jalloh I, Helmy A, Guilfoyle MR, Grice P, Mason A, Giorgi-Coll S, Gallagher CN, Murphy MP, Menon DK, Carpenter TA, Hutchinson PJ, Carpenter KLH. High-physiological and supra-physiological 1,2- 13C 2 glucose focal supplementation to the traumatised human brain. J Cereb Blood Flow Metab 2023; 43:1685-1701. [PMID: 37157814 PMCID: PMC10581237 DOI: 10.1177/0271678x231173584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 03/12/2023] [Accepted: 04/02/2023] [Indexed: 05/10/2023]
Abstract
How to optimise glucose metabolism in the traumatised human brain remains unclear, including whether injured brain can metabolise additional glucose when supplied. We studied the effect of microdialysis-delivered 1,2-13C2 glucose at 4 and 8 mmol/L on brain extracellular chemistry using bedside ISCUSflex, and the fate of the 13C label in the 8 mmol/L group using high-resolution NMR of recovered microdialysates, in 20 patients. Compared with unsupplemented perfusion, 4 mmol/L glucose increased extracellular concentrations of pyruvate (17%, p = 0.04) and lactate (19%, p = 0.01), with a small increase in lactate/pyruvate ratio (5%, p = 0.007). Perfusion with 8 mmol/L glucose did not significantly influence extracellular chemistry measured with ISCUSflex, compared to unsupplemented perfusion. These extracellular chemistry changes appeared influenced by the underlying metabolic states of patients' traumatised brains, and the presence of relative neuroglycopaenia. Despite abundant 13C glucose supplementation, NMR revealed only 16.7% 13C enrichment of recovered extracellular lactate; the majority being glycolytic in origin. Furthermore, no 13C enrichment of TCA cycle-derived extracellular glutamine was detected. These findings indicate that a large proportion of extracellular lactate does not originate from local glucose metabolism, and taken together with our earlier studies, suggest that extracellular lactate is an important transitional step in the brain's production of glutamine.
Collapse
Affiliation(s)
- Matthew G Stovell
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Department of Neurosurgery, The Walton Centre, Liverpool, UK
| | - Duncan J Howe
- Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Eric P Thelin
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Ibrahim Jalloh
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Adel Helmy
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Mathew R Guilfoyle
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Peter Grice
- Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Andrew Mason
- Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Susan Giorgi-Coll
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Clare N Gallagher
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Calgary, Calgary, Canada
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - David K Menon
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - T Adrian Carpenter
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Peter J Hutchinson
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Keri LH Carpenter
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
8
|
Muñoz-Ballester C, Robel S. Astrocyte-mediated mechanisms contribute to traumatic brain injury pathology. WIREs Mech Dis 2023; 15:e1622. [PMID: 37332001 PMCID: PMC10526985 DOI: 10.1002/wsbm.1622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 05/25/2023] [Accepted: 05/29/2023] [Indexed: 06/20/2023]
Abstract
Astrocytes respond to traumatic brain injury (TBI) with changes to their molecular make-up and cell biology, which results in changes in astrocyte function. These changes can be adaptive, initiating repair processes in the brain, or detrimental, causing secondary damage including neuronal death or abnormal neuronal activity. The response of astrocytes to TBI is often-but not always-accompanied by the upregulation of intermediate filaments, including glial fibrillary acidic protein (GFAP) and vimentin. Because GFAP is often upregulated in the context of nervous system disturbance, reactive astrogliosis is sometimes treated as an "all-or-none" process. However, the extent of astrocytes' cellular, molecular, and physiological adjustments is not equal for each TBI type or even for each astrocyte within the same injured brain. Additionally, new research highlights that different neurological injuries and diseases result in entirely distinctive and sometimes divergent astrocyte changes. Thus, extrapolating findings on astrocyte biology from one pathological context to another is problematic. We summarize the current knowledge about astrocyte responses specific to TBI and point out open questions that the field should tackle to better understand how astrocytes shape TBI outcomes. We address the astrocyte response to focal versus diffuse TBI and heterogeneity of reactive astrocytes within the same brain, the role of intermediate filament upregulation, functional changes to astrocyte function including potassium and glutamate homeostasis, blood-brain barrier maintenance and repair, metabolism, and reactive oxygen species detoxification, sex differences, and factors influencing astrocyte proliferation after TBI. This article is categorized under: Neurological Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Carmen Muñoz-Ballester
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Stefanie Robel
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
9
|
Omori NE, Woo GH, Mansor LS. Exogenous Ketones and Lactate as a Potential Therapeutic Intervention for Brain Injury and Neurodegenerative Conditions. Front Hum Neurosci 2022; 16:846183. [PMID: 36267349 PMCID: PMC9577611 DOI: 10.3389/fnhum.2022.846183] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Metabolic dysfunction is a ubiquitous underlying feature of many neurological conditions including acute traumatic brain injuries and chronic neurodegenerative conditions. A central problem in neurological patients, in particular those with traumatic brain injuries, is an impairment in the utilization of glucose, which is the predominant metabolic substrate in a normally functioning brain. In such patients, alternative substrates including ketone bodies and lactate become important metabolic candidates for maintaining brain function. While the potential neuroprotective benefits of ketosis have been recognized for up to almost a century, the majority of work has focused on the use of ketogenic diets to induce such a state, which is inappropriate in cases of acute disease due to the prolonged periods of time (i.e., weeks to months) required for the effects of a ketogenic diet to be seen. The following review seeks to explore the neuroprotective effects of exogenous ketone and lactate preparations, which have more recently become commercially available and are able to induce a deep ketogenic response in a fraction of the time. The rapid response of exogenous preparations makes their use as a therapeutic adjunct more feasible from a clinical perspective in both acute and chronic neurological conditions. Potentially, their ability to globally moderate long-term, occult brain dysfunction may also be relevant in reducing lifetime risks of certain neurodegenerative conditions. In particular, this review explores the association between traumatic brain injury and contusion-related dementia, assessing metabolic parallels and highlighting the potential role of exogenous ketone and lactate therapies.
Collapse
|
10
|
Serpa RO, Ferguson L, Larson C, Bailard J, Cooke S, Greco T, Prins ML. Pathophysiology of Pediatric Traumatic Brain Injury. Front Neurol 2021; 12:696510. [PMID: 34335452 PMCID: PMC8319243 DOI: 10.3389/fneur.2021.696510] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/21/2021] [Indexed: 11/23/2022] Open
Abstract
The national incidence of traumatic brain injury (TBI) exceeds that of any other disease in the pediatric population. In the United States the Centers for Disease Control and Prevention (CDC) reports 697,347 annual TBIs in children ages 0–19 that result in emergency room visits, hospitalization or deaths. There is a bimodal distribution within the pediatric TBI population, with peaks in both toddlers and adolescents. Preclinical TBI research provides evidence for age differences in acute pathophysiology that likely contribute to long-term outcome differences between age groups. This review will examine the timecourse of acute pathophysiological processes during cerebral maturation, including calcium accumulation, glucose metabolism and cerebral blood flow. Consequences of pediatric TBI are complicated by the ongoing maturational changes allowing for substantial plasticity and windows of vulnerabilities. This review will also examine the timecourse of later outcomes after mild, repeat mild and more severe TBI to establish developmental windows of susceptibility and altered maturational trajectories. Research progress for pediatric TBI is critically important to reveal age-associated mechanisms and to determine knowledge gaps for future studies.
Collapse
Affiliation(s)
- Rebecka O Serpa
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lindsay Ferguson
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Cooper Larson
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Julie Bailard
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Samantha Cooke
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Tiffany Greco
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Mayumi L Prins
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
11
|
Liang J, Han R, Zhou B. Metabolic Reprogramming: Strategy for Ischemic Stroke Treatment by Ischemic Preconditioning. BIOLOGY 2021; 10:biology10050424. [PMID: 34064579 PMCID: PMC8151271 DOI: 10.3390/biology10050424] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 01/15/2023]
Abstract
Stroke is one of the leading causes of death and permanent disability worldwide. Ischemic preconditioning (IPC) is an endogenous protective strategy, which has been reported to exhibit a significant neuroprotective effect in reducing the incidence of ischemic stroke. However, the underlying neuroprotective mechanisms of IPC remain elusive. An increased understanding of the pathogenic mechanisms of stroke and IPC serves to highlight the importance of metabolic reprogramming. In this review, we summarize the metabolic disorder and metabolic plasticity in the incidence and progression of ischemic stroke. We also elaborate how IPC fully mobilizes the metabolic reprogramming to maintain brain metabolic homeostasis, especially for energy and redox homeostasis, and finally protects brain function in the event of an ischemic stroke.
Collapse
Affiliation(s)
- Jing Liang
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Interdisciplinary Innovation Institute of Medicine and Engineering, Beihang University, Beijing 100191, China; (J.L.); (R.H.)
| | - Rongrong Han
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Interdisciplinary Innovation Institute of Medicine and Engineering, Beihang University, Beijing 100191, China; (J.L.); (R.H.)
| | - Bing Zhou
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Interdisciplinary Innovation Institute of Medicine and Engineering, Beihang University, Beijing 100191, China; (J.L.); (R.H.)
- School of Engineering Medicine, Beihang University, Beijing 100191, China
- Correspondence:
| |
Collapse
|
12
|
Takahashi CE, Virmani D, Chung DY, Ong C, Cervantes-Arslanian AM. Blunt and Penetrating Severe Traumatic Brain Injury. Neurol Clin 2021; 39:443-469. [PMID: 33896528 DOI: 10.1016/j.ncl.2021.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Severe traumatic brain injury is a common problem. Current practices focus on the importance of early resuscitation, transfer to high-volume centers, and provider expertise across multiple specialties. In the emergency department, patients should receive urgent intracranial imaging and consideration for tranexamic acid. Close observation in the intensive care unit environment helps identify problems, such as seizure, intracranial pressure crisis, and injury progression. In addition to traditional neurologic examination, patients benefit from use of intracranial monitors. Monitors gather physiologic data on intracranial and cerebral perfusion pressures to help guide therapy. Brain tissue oxygenation monitoring and cerebromicrodialysis show promise in studies.
Collapse
Affiliation(s)
- Courtney E Takahashi
- Department of Neurology, Boston Medical Center, 72 East Concord Street, Collamore, C-3, Boston, MA 02118, USA.
| | - Deepti Virmani
- Department of Neurology, Boston University School of Medicine and Boston Medical Center, 72 East Concord Street, Collamore, C-3, Boston, MA 02118, USA
| | - David Y Chung
- Department of Neurology, Boston University School of Medicine and Boston Medical Center, 72 East Concord Street, Collamore, C-3, Boston, MA 02118, USA; Division of Neurocritical Care, Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA; Neurovascular Research Unit, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Charlene Ong
- Department of Neurology, Boston University School of Medicine and Boston Medical Center, 72 East Concord Street, Collamore, C-3, Boston, MA 02118, USA
| | - Anna M Cervantes-Arslanian
- Boston University School of Medicine and Boston Medical Center, 72 East Concord Street, Collamore, C-3, Boston, MA 02118, USA
| |
Collapse
|
13
|
Xu XJ, Yang MS, Zhang B, Niu F, Dong JQ, Liu BY. Glucose metabolism: A link between traumatic brain injury and Alzheimer's disease. Chin J Traumatol 2021; 24:5-10. [PMID: 33358332 PMCID: PMC7878452 DOI: 10.1016/j.cjtee.2020.10.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 10/08/2020] [Accepted: 10/15/2020] [Indexed: 02/04/2023] Open
Abstract
Traumatic brain injury (TBI), a growing public health problem, is a leading cause of death and disability worldwide, although its prevention measures and clinical cares are substantially improved. Increasing evidence shows that TBI may increase the risk of mood disorders and neurodegenerative diseases, including Alzheimer's disease (AD). However, the complex relationship between TBI and AD remains elusive. Metabolic dysfunction has been the common pathology in both TBI and AD. On the one hand, TBI perturbs the glucose metabolism of the brain, and causes energy crisis and subsequent hyperglycolysis. On the other hand, glucose deprivation promotes amyloidogenesis via β-site APP cleaving enzyme-1 dependent mechanism, and triggers tau pathology and synaptic function. Recent findings suggest that TBI might facilitate Alzheimer's pathogenesis by altering metabolism, which provides clues to metabolic link between TBI and AD. In this review, we will explore how TBI-induced metabolic changes contribute to the development of AD.
Collapse
Affiliation(s)
- Xiao-Jian Xu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Meng-Shi Yang
- Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Bin Zhang
- Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Fei Niu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
| | - Jin-Qian Dong
- Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Bai-Yun Liu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China,Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China,Nerve Injury and Repair Center of Beijing Institute for Brain Disorders, Beijing, 100070, China,China National Clinical Research Center for Neurological Diseases, Beijing, 100070, China,Corresponding author. Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100070, China.
| |
Collapse
|
14
|
Shi AC, Rohlwink U, Scafidi S, Kannan S. Microglial Metabolism After Pediatric Traumatic Brain Injury - Overlooked Bystanders or Active Participants? Front Neurol 2021; 11:626999. [PMID: 33569038 PMCID: PMC7868439 DOI: 10.3389/fneur.2020.626999] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 12/23/2020] [Indexed: 12/14/2022] Open
Abstract
Microglia play an integral role in brain development but are also crucial for repair and recovery after traumatic brain injury (TBI). TBI induces an intense innate immune response in the immature, developing brain that is associated with acute and chronic changes in microglial function. These changes contribute to long-lasting consequences on development, neurologic function, and behavior. Although alterations in glucose metabolism are well-described after TBI, the bulk of the data is focused on metabolic alterations in astrocytes and neurons. To date, the interplay between alterations in intracellular metabolic pathways in microglia and the innate immune response in the brain following an injury is not well-studied. In this review, we broadly discuss the microglial responses after TBI. In addition, we highlight reported metabolic alterations in microglia and macrophages, and provide perspective on how changes in glucose, fatty acid, and amino acid metabolism can influence and modulate the microglial phenotype and response to injury.
Collapse
Affiliation(s)
- Aria C Shi
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ursula Rohlwink
- Neuroscience Institute and Division of Neurosurgery, University of Cape Town, Cape Town, South Africa.,The Francis Crick Institute, London, United Kingdom
| | - Susanna Scafidi
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
15
|
Cherkas A, Holota S, Mdzinarashvili T, Gabbianelli R, Zarkovic N. Glucose as a Major Antioxidant: When, What for and Why It Fails? Antioxidants (Basel) 2020; 9:antiox9020140. [PMID: 32033390 PMCID: PMC7070274 DOI: 10.3390/antiox9020140] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/01/2020] [Accepted: 02/03/2020] [Indexed: 02/07/2023] Open
Abstract
A human organism depends on stable glucose blood levels in order to maintain its metabolic needs. Glucose is considered to be the most important energy source, and glycolysis is postulated as a backbone pathway. However, when the glucose supply is limited, ketone bodies and amino acids can be used to produce enough ATP. In contrast, for the functioning of the pentose phosphate pathway (PPP) glucose is essential and cannot be substituted by other metabolites. The PPP generates and maintains the levels of nicotinamide adenine dinucleotide phosphate (NADPH) needed for the reduction in oxidized glutathione and protein thiols, the synthesis of lipids and DNA as well as for xenobiotic detoxification, regulatory redox signaling and counteracting infections. The flux of glucose into a PPP—particularly under extreme oxidative and toxic challenges—is critical for survival, whereas the glycolytic pathway is primarily activated when glucose is abundant, and there is lack of NADP+ that is required for the activation of glucose-6 phosphate dehydrogenase. An important role of glycogen stores in resistance to oxidative challenges is discussed. Current evidences explain the disruptive metabolic effects and detrimental health consequences of chronic nutritional carbohydrate overload, and provide new insights into the positive metabolic effects of intermittent fasting, caloric restriction, exercise, and ketogenic diet through modulation of redox homeostasis.
Collapse
Affiliation(s)
- Andriy Cherkas
- Department of Internal Medicine # 1, Lviv National Medical University, 79010 Lviv, Ukraine
- Correspondence:
| | - Serhii Holota
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Lviv National Medical University, 79010 Lviv, Ukraine;
- Department of Organic Chemistry and Pharmacy, Lesya Ukrainka Eastern European National University, 43025 Lutsk, Ukraine
| | - Tamaz Mdzinarashvili
- Institute of Medical and Applied Biophysics, I. Javakhishvili Tbilisi State University, 0128 Tbilisi, Georgia;
| | - Rosita Gabbianelli
- Unit of Molecular Biology, School of Pharmacy, University of Camerino, 62032 Camerino, Italy;
| | - Neven Zarkovic
- Laboratory for Oxidative Stress (LabOS), Institute “Rudjer Boskovic”, HR-10000 Zagreb, Croatia;
| |
Collapse
|
16
|
The behavioural and pathophysiological effects of the ketogenic diet on mild traumatic brain injury in adolescent rats. Behav Brain Res 2019; 376:112225. [DOI: 10.1016/j.bbr.2019.112225] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/16/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023]
|
17
|
Sugar as a therapeutic target for the cognitive restoration following traumatic brain injury. Curr Opin Neurol 2019; 32:815-821. [PMID: 31609736 DOI: 10.1097/wco.0000000000000752] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW This review aims to discuss examples of changes in glucose (sugar) metabolism after traumatic brain injury (TBI). It will attempt to provide an understanding of what changes in glucose metabolism mean for the injured brain. It will further identify potential therapeutic target(s) emanating from our growing understanding of glucose pathways and their roles in TBI. RECENT FINDINGS Although a significant fraction of glucose is utilized for the energy production in the brain, a small fraction is utilized in other, often ignored pathways. Recent studies have unraveled unexpected biological effects of glucose through these pathways, including redox regulation, genetic and epigenetic regulation, glycation of proteins, nucleotide synthesis and amino acid synthesis. SUMMARY A number of regulatory players in minor glucose metabolic pathways, such as folate and chondroitin sulfate proteoglycans, have recently been identified as potential targets to restore cognitive functions. Targeting of these players should be combined with the supplementation of alternative energy substrates to achieve the maximal cognitive restoration after TBI. This multimodal therapeutic strategy deserves testing in various models of TBI. VIDEO ABSTRACT Supplemental digital video content 1: Video that demonstrates an effective therapeutic strategy for the cognitive restoration after TBI. http://links.lww.com/CONR/A46.
Collapse
|
18
|
Bordone MP, Salman MM, Titus HE, Amini E, Andersen JV, Chakraborti B, Diuba AV, Dubouskaya TG, Ehrke E, Espindola de Freitas A, Braga de Freitas G, Gonçalves RA, Gupta D, Gupta R, Ha SR, Hemming IA, Jaggar M, Jakobsen E, Kumari P, Lakkappa N, Marsh APL, Mitlöhner J, Ogawa Y, Paidi RK, Ribeiro FC, Salamian A, Saleem S, Sharma S, Silva JM, Singh S, Sulakhiya K, Tefera TW, Vafadari B, Yadav A, Yamazaki R, Seidenbecher CI. The energetic brain - A review from students to students. J Neurochem 2019; 151:139-165. [PMID: 31318452 DOI: 10.1111/jnc.14829] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 07/03/2019] [Accepted: 07/08/2019] [Indexed: 12/12/2022]
Abstract
The past 20 years have resulted in unprecedented progress in understanding brain energy metabolism and its role in health and disease. In this review, which was initiated at the 14th International Society for Neurochemistry Advanced School, we address the basic concepts of brain energy metabolism and approach the question of why the brain has high energy expenditure. Our review illustrates that the vertebrate brain has a high need for energy because of the high number of neurons and the need to maintain a delicate interplay between energy metabolism, neurotransmission, and plasticity. Disturbances to the energetic balance, to mitochondria quality control or to glia-neuron metabolic interaction may lead to brain circuit malfunction or even severe disorders of the CNS. We cover neuronal energy consumption in neural transmission and basic ('housekeeping') cellular processes. Additionally, we describe the most common (glucose) and alternative sources of energy namely glutamate, lactate, ketone bodies, and medium chain fatty acids. We discuss the multifaceted role of non-neuronal cells in the transport of energy substrates from circulation (pericytes and astrocytes) and in the supply (astrocytes and microglia) and usage of different energy fuels. Finally, we address pathological consequences of disrupted energy homeostasis in the CNS.
Collapse
Affiliation(s)
- Melina Paula Bordone
- Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mootaz M Salman
- Department of Cell Biology, Harvard Medical School, and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Haley E Titus
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Elham Amini
- Department of Medicine, University Kebangsaan Malaysia Medical Centre (HUKM), Cheras, Kuala Lumpur, Malaysia
| | - Jens V Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Artem V Diuba
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Tatsiana G Dubouskaya
- Institute of Biophysics and Cell Engineering, National Academy of Sciences of Belarus, Minsk, Belarus
| | - Eric Ehrke
- Centre for Biomolecular Interactions, University of Bremen, Bremen, Germany
| | - Andiara Espindola de Freitas
- Neurobiology Section, Biological Sciences Division, University of California, San Diego, La Jolla, California, USA
| | | | | | | | - Richa Gupta
- CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Sharon R Ha
- Baylor College of Medicine, Houston, Texas, USA
| | - Isabel A Hemming
- Brain Growth and Disease Laboratory, The Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia.,School of Medicine and Pharmacology, The University of Western Australia, Crawley, Australia
| | - Minal Jaggar
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Emil Jakobsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Punita Kumari
- Defense Institute of Physiology and allied sciences, Defense Research and Development Organization, Timarpur, Delhi, India
| | - Navya Lakkappa
- Department of Pharmacology, JSS college of Pharmacy, Ooty, India
| | - Ashley P L Marsh
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Jessica Mitlöhner
- Department of Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology Magdeburg, Magdeburg, Germany
| | - Yuki Ogawa
- The Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| | | | | | - Ahmad Salamian
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Suraiya Saleem
- CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Sorabh Sharma
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Rajasthan, India
| | - Joana M Silva
- Life and Health Sciences Research Institute (ICVS), Medical School, University of Minho, Braga, Portugal
| | - Shripriya Singh
- CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Kunjbihari Sulakhiya
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, India
| | - Tesfaye Wolde Tefera
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Behnam Vafadari
- Institute of environmental medicine, UNIKA-T, Technical University of Munich, Munich, Germany
| | - Anuradha Yadav
- CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Reiji Yamazaki
- Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan.,Center for Behavioral Brain Sciences (CBBS), Otto von Guericke University, Magdeburg, Germany
| | - Constanze I Seidenbecher
- Department of Neurochemistry and Molecular Biology, Leibniz Institute for Neurobiology Magdeburg, Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Otto von Guericke University, Magdeburg, Germany
| |
Collapse
|
19
|
Emery Joseph Crownover J, Holland AM. Therapeutic ketosis for mild traumatic brain injury. TRANSLATIONAL SPORTS MEDICINE 2019. [DOI: 10.1002/tsm2.89] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
| | - Angelia Maleah Holland
- Nutrition, Exercise, and Stress Laboratory, Department of Kinesiology Augusta University Augusta Georgia
| |
Collapse
|
20
|
Agoston DV, Vink R, Helmy A, Risling M, Nelson D, Prins M. How to Translate Time: The Temporal Aspects of Rodent and Human Pathobiological Processes in Traumatic Brain Injury. J Neurotrauma 2019; 36:1724-1737. [PMID: 30628544 PMCID: PMC7643768 DOI: 10.1089/neu.2018.6261] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Traumatic brain injury (TBI) triggers multiple pathobiological responses with differing onsets, magnitudes, and durations. Identifying the therapeutic window of individual pathologies is critical for successful pharmacological treatment. Dozens of experimental pharmacotherapies have been successfully tested in rodent models, yet all of them (to date) have failed in clinical trials. The differing time scales of rodent and human biological and pathological processes may have contributed to these failures. We compared rodent versus human time scales of TBI-induced changes in cerebral glucose metabolism, inflammatory processes, axonal integrity, and water homeostasis based on published data. We found that the trajectories of these pathologies run on different timescales in the two species, and it appears that there is no universal "conversion rate" between rodent and human pathophysiological processes. For example, the inflammatory process appears to have an abbreviated time scale in rodents versus humans relative to cerebral glucose metabolism or axonal pathologies. Limitations toward determining conversion rates for various pathobiological processes include the use of differing outcome measures in experimental and clinical TBI studies and the rarity of longitudinal studies. In order to better translate time and close the translational gap, we suggest 1) using clinically relevant outcome measures, primarily in vivo imaging and blood-based proteomics, in experimental TBI studies and 2) collecting data at multiple post-injury time points with a frequency exceeding the expected information content by two or three times. Combined with a big data approach, we believe these measures will facilitate the translation of promising experimental treatments into clinical use.
Collapse
Affiliation(s)
- Denes V. Agoston
- Department of Anatomy, Physiology and Genetics, Uniformed Services University, Bethesda, Maryland
| | - Robert Vink
- Division of Health Science, University of South Australia, Adelaide, Australia
| | - Adel Helmy
- Division of Neurosurgery, Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Mårten Risling
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - David Nelson
- Department of Physiology and Pharmacology, Section of Perioperative Medicine and Intensive Care, Karolinska Institutet, Stockholm, Sweden
| | - Mayumi Prins
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, California
| |
Collapse
|
21
|
Chitturi J, Li Y, Santhakumar V, Kannurpatti SS. Consolidated Biochemical Profile of Subacute Stage Traumatic Brain Injury in Early Development. Front Neurosci 2019; 13:431. [PMID: 31130841 PMCID: PMC6509949 DOI: 10.3389/fnins.2019.00431] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 04/15/2019] [Indexed: 11/30/2022] Open
Abstract
Traumatic brain injury (TBI) in general has varied neuropathological consequences depending upon the intensity and biomechanics of the injury. Furthermore, in pediatric TBI, intrinsic developmental changes add further complexity, necessitating a biochemical dimension for improved TBI characterization. In our earlier study investigating the subacute stage TBI metabolome (72 h post-injury) in a developmental rat model, significant ipsilateral brain biochemical changes occurred across 25 metabolite sets as determined by metabolite set enrichment analysis (MSEA). The broad metabolic perturbation was accompanied by behavioral deficits and neuronal loss across the ipsilateral hemisphere containing the injury epicenter. In order to obtain a consolidated biochemical profile of the TBI assessment, a subgrouping of the 190 identified brain metabolites was performed. Metabolites were divided into seven major subgroups: oxidative energy/mitochondrial, glycolysis/pentose phosphate pathway, fatty acid, amino acid, neurotransmitters/neuromodulators, one-carbon/folate and other metabolites. Subgroups were based on the chemical nature and association with critically altered biochemical pathways after TBI as obtained from our earlier untargeted analysis. Each metabolite subgroup extracted from the ipsilateral sham and TBI brains were modeled using multivariate partial least square discriminant analysis (PLS-DA) with the model accuracy used as a measurable index of TBI neurochemical impact. Volcano plots of each subgroup, corrected for multiple comparisons, determined the TBI neurochemical specificity. The results provide a ranked biochemical profile along with specificity of changes after developmental TBI, enabling a consolidated biochemical template for future classification of different TBI intensities and injury types in animal models.
Collapse
Affiliation(s)
- Jyothsna Chitturi
- Department of Radiology, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ, United States
| | - Ying Li
- Department of Pharmacology, Physiology & Neuroscience, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ, United States
| | - Vijayalakshmi Santhakumar
- Department of Pharmacology, Physiology & Neuroscience, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ, United States.,Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Sridhar S Kannurpatti
- Department of Radiology, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ, United States
| |
Collapse
|
22
|
Sta Maria NS, Sargolzaei S, Prins ML, Dennis EL, Asarnow RF, Hovda DA, Harris NG, Giza CC. Bridging the gap: Mechanisms of plasticity and repair after pediatric TBI. Exp Neurol 2019; 318:78-91. [PMID: 31055004 DOI: 10.1016/j.expneurol.2019.04.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 03/09/2019] [Accepted: 04/25/2019] [Indexed: 01/25/2023]
Abstract
Traumatic brain injury is the leading cause of death and disability in the United States, and may be associated with long lasting impairments into adulthood. The multitude of ongoing neurobiological processes that occur during brain maturation confer both considerable vulnerability to TBI but may also provide adaptability and potential for recovery. This review will examine and synthesize our current understanding of developmental neurobiology in the context of pediatric TBI. Delineating this biology will facilitate more targeted initial care, mechanism-based therapeutic interventions and better long-term prognostication and follow-up.
Collapse
Affiliation(s)
- Naomi S Sta Maria
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, University of Southern California, 1501 San Pablo Street, ZNI115, Los Angeles, CA 90033, United States of America.
| | - Saman Sargolzaei
- UCLA Brain Injury Research Center, Department of Neurosurgery, University of California at Los Angeles, Box 956901, 300 Stein Plaza, Ste 562, 5th Floor, Los Angeles, CA 90095-6901, United States of America.
| | - Mayumi L Prins
- UCLA Brain Injury Research Center, Department of Neurosurgery, University of California at Los Angeles, Box 956901, 300 Stein Plaza, Ste 562, 5th Floor, Los Angeles, CA 90095-6901, United States of America; Steve Tisch BrainSPORT Program, University of California at Los Angeles, Los Angeles, CA, United States of America.
| | - Emily L Dennis
- Brigham and Women's Hospital/Harvard University and Department of Psychology, Stanford University, 1249 Boylston Street, Boston, MA 02215, United States of America.
| | - Robert F Asarnow
- Department of Psychiatry and Biobehavioral Sciences, University of California at Los Angeles, Box 951759, 760 Westwood Plaza, 48-240C Semel Institute, Los Angeles, CA 90095-1759, United States of America.
| | - David A Hovda
- UCLA Brain Injury Research Center, Department of Neurosurgery, University of California at Los Angeles, Box 956901, 300 Stein Plaza, Ste 562, 5th Floor, Los Angeles, CA 90095-6901, United States of America; Department of Medical and Molecular Pharmacology, University of California at Los Angeles, Box 956901, 300 Stein Plaza, Ste 562 & Semel 18-228A, Los Angeles, CA 90095-6901, United States of America.
| | - Neil G Harris
- UCLA Brain Injury Research Center, Department of Neurosurgery, University of California at Los Angeles, Box 956901, 300 Stein Plaza, Ste 562, 5th Floor, Los Angeles, CA 90095-6901, United States of America; Intellectual Development and Disabilities Research Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, United States of America.
| | - Christopher C Giza
- UCLA Brain Injury Research Center, Department of Neurosurgery, University of California at Los Angeles, Box 956901, 300 Stein Plaza, Ste 562, 5th Floor, Los Angeles, CA 90095-6901, United States of America; Steve Tisch BrainSPORT Program, University of California at Los Angeles, Los Angeles, CA, United States of America; Division of Pediatric Neurology, Mattel Children's Hospital - UCLA, Los Angeles, CA, United States of America.
| |
Collapse
|
23
|
McConnell HL, Li Z, Woltjer RL, Mishra A. Astrocyte dysfunction and neurovascular impairment in neurological disorders: Correlation or causation? Neurochem Int 2019; 128:70-84. [PMID: 30986503 DOI: 10.1016/j.neuint.2019.04.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 12/14/2022]
Abstract
The neurovascular unit, consisting of neurons, astrocytes, and vascular cells, has become the focus of much discussion in the last two decades and emerging literature now suggests an association between neurovascular dysfunction and neurological disorders. In this review, we synthesize the known and suspected contributions of astrocytes to neurovascular dysfunction in disease. Throughout the brain, astrocytes are centrally positioned to dynamically mediate interactions between neurons and the cerebral vasculature, and play key roles in blood-brain barrier maintenance and neurovascular coupling. It is increasingly apparent that the changes in astrocytes in response to a variety of insults to brain tissue -collectively referred to as "reactive astrogliosis" - are not just an epiphenomenon restricted to morphological alterations, but comprise functional changes in astrocytes that contribute to the phenotype of neurological diseases with both beneficial and detrimental effects. In the context of the neurovascular unit, astrocyte dysfunction accompanies, and may contribute to, blood-brain barrier impairment and neurovascular dysregulation, highlighting the need to determine the exact nature of the relationship between astrocyte dysfunction and neurovascular impairments. Targeting astrocytes may represent a new strategy in combinatorial therapeutics for preventing the mismatch of energy supply and demand that often accompanies neurological disorders.
Collapse
Affiliation(s)
- Heather L McConnell
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, United States
| | - Zhenzhou Li
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, United States; Department of Anesthesiology, General Hospital of Ningxia Medical University, Yinchuan City, China
| | - Randall L Woltjer
- Department of Neuropathology, Oregon Health & Science University, Portland, OR, United States
| | - Anusha Mishra
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, United States.
| |
Collapse
|
24
|
Metabolic perturbations after pediatric TBI: It's not just about glucose. Exp Neurol 2019; 316:74-84. [PMID: 30951705 DOI: 10.1016/j.expneurol.2019.03.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/13/2019] [Accepted: 03/30/2019] [Indexed: 12/22/2022]
Abstract
Improved patient survival following pediatric traumatic brain injury (TBI) has uncovered a currently limited understanding of both the adaptive and maladaptive metabolic perturbations that occur during the acute and long-term phases of recovery. While much is known about the redundancy of metabolic pathways that provide adequate energy and substrates for normal brain growth and development, the field is only beginning to characterize perturbations in these metabolic pathways after pediatric TBI. To date, the majority of studies have focused on dysregulated oxidative glucose metabolism after injury; however, the immature brain is well-equipped to use alternative substrates to fuel energy production, growth, and development. A comprehensive understanding of metabolic changes associated with pediatric TBI cannot be limited to investigations of glucose metabolism alone. All energy substrates used by the brain should be considered in developing nutritional and pharmacological interventions for pediatric head trauma. This review summarizes post-injury changes in brain metabolism of glucose, lipids, ketone bodies, and amino acids with discussion of the therapeutic potential of altering substrate utilization to improve pediatric TBI outcomes.
Collapse
|
25
|
Gonçalves CA, Rodrigues L, Bobermin LD, Zanotto C, Vizuete A, Quincozes-Santos A, Souza DO, Leite MC. Glycolysis-Derived Compounds From Astrocytes That Modulate Synaptic Communication. Front Neurosci 2019; 12:1035. [PMID: 30728759 PMCID: PMC6351787 DOI: 10.3389/fnins.2018.01035] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 12/20/2018] [Indexed: 12/14/2022] Open
Abstract
Based on the concept of the tripartite synapse, we have reviewed the role of glucose-derived compounds in glycolytic pathways in astroglial cells. Glucose provides energy and substrate replenishment for brain activity, such as glutamate and lipid synthesis. In addition, glucose metabolism in the astroglial cytoplasm results in products such as lactate, methylglyoxal, and glutathione, which modulate receptors and channels in neurons. Glucose has four potential destinations in neural cells, and it is possible to propose a crossroads in “X” that can be used to describe these four destinations. Glucose-6P can be used either for glycogen synthesis or the pentose phosphate pathway on the left and right arms of the X, respectively. Fructose-6P continues through the glycolysis pathway until pyruvate is formed but can also act as the initial compound in the hexosamine pathway, representing the left and right legs of the X, respectively. We describe each glucose destination and its regulation, indicating the products of these pathways and how they can affect synaptic communication. Extracellular L-lactate, either generated from glucose or from glycogen, binds to HCAR1, a specific receptor that is abundantly localized in perivascular and post-synaptic membranes and regulates synaptic plasticity. Methylglyoxal, a product of a deviation of glycolysis, and its derivative D-lactate are also released by astrocytes and bind to GABAA receptors and HCAR1, respectively. Glutathione, in addition to its antioxidant role, also binds to ionotropic glutamate receptors in the synaptic cleft. Finally, we examined the hexosamine pathway and evaluated the effect of GlcNAc-modification on key proteins that regulate the other glucose destinations.
Collapse
Affiliation(s)
- Carlos-Alberto Gonçalves
- Department of Biochemistry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Letícia Rodrigues
- Department of Biochemistry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Larissa D Bobermin
- Department of Biochemistry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Caroline Zanotto
- Department of Biochemistry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Adriana Vizuete
- Department of Biochemistry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - André Quincozes-Santos
- Department of Biochemistry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Diogo O Souza
- Department of Biochemistry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Marina C Leite
- Department of Biochemistry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| |
Collapse
|
26
|
|
27
|
Chae MK, Lee SE, Kang SY, Sim MS. Monitoring of Cerebral Metabolism in Postcardiac Arrest Patients: A Pilot Study. Ther Hypothermia Temp Manag 2018; 8:234-238. [PMID: 30124387 PMCID: PMC6302816 DOI: 10.1089/ther.2018.0018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
For better care of postcardiac arrest patients, objective serial assessments of brain injury severity are needed. We hypothesized that monitoring of cerebral energy metabolism based on arterio-jugular (AJ) differences of metabolites will provide serial details of brain injury and information about neurologic outcomes in patients. Measurements of lactate and glucose in addition to blood gas analyses were done every 6 hours from the radial artery and jugular bulb in postcardiac arrest patients throughout targeted temperature management (TTM). Jugular bulb saturation, AJ difference of O2, and AJ difference of lactate (AJDL) were calculated and compared between the different neurologic outcome groups. Linear mixed-model analysis was done to assess AJDL based on the different phases of TTM and neurologic outcome. A total of 13 patients were included in the study (n = 4 good outcome, n = 9 poor outcome). AJDL as an indicator of cerebral metabolism was significantly different between the outcome groups and demonstrated negative values in the poor neurologic outcome group (0.06 [0.05-0.09] vs. -0.14 [-0.06 to -0.27], p < 0.01). However, there was no significant difference in AJDL between the outcome groups in the mixed effects model (p = 0.05). In addition, there were no differences between the phases of TTM in both groups (p = 0.46). AJDL was observed to be informative but was not significantly different between neurologic outcome groups throughout the different phases of TTM in our pilot study. Future studies are needed for further investigation of AJDL as an indicator of brain injury severity.
Collapse
Affiliation(s)
- Minjung Kathy Chae
- Department of Emergency Medicine, Ajou University School of Medicine, Suwon, Korea
| | - Sung Eun Lee
- Department of Emergency Medicine, Ajou University School of Medicine, Suwon, Korea.,Department of Neurology, Ajou University School of Medicine, Suwon, Korea
| | - So Young Kang
- Office of Biostatistics, Institute of Medical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Min Seob Sim
- Department of Emergency Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
28
|
Wolahan SM, Lebby E, Mao HC, McArthur D, Real C, Vespa P, Braas D, Glenn TC. Novel Metabolomic Comparison of Arterial and Jugular Venous Blood in Severe Adult Traumatic Brain Injury Patients and the Impact of Pentobarbital Infusion. J Neurotrauma 2018; 36:212-221. [PMID: 29901425 DOI: 10.1089/neu.2018.5674] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Treatment of severe traumatic brain injury (TBI) in the intensive care unit focuses on controlling intracranial pressure, ensuring sufficient cerebral perfusion, and monitoring for secondary injuries. However, there are limited prognostic tools and no biomarkers or tests of the evolving neuropathology. Metabolomics has the potential to be a powerful tool to indirectly monitor evolving dysfunctional metabolism. We compared metabolite levels in simultaneously collected arterial and jugular venous samples in acute TBI patients undergoing intensive care as well as in healthy control volunteers. Our results show that, first, many circulating metabolites are decreased in TBI patients compared with healthy controls days after injury; both proline and hydroxyproline were depleted by ≥60% compared with healthy controls, as was gluconate. Second, both arterial and jugular venous plasma metabolomic analysis separates TBI patients from healthy controls and shows that distinct combinations of metabolites are driving the group separation in the two blood types. Third, TBI patients under heavy sedation with pentobarbital at the time of blood collection were discernibly different from patients not receiving pentobarbital. These results highlight the importance of accounting for medications in metabolomics analysis. Jugular venous plasma metabolomics shows potential as a minimally invasive tool to identify and study dysfunctional cerebral metabolism after TBI.
Collapse
Affiliation(s)
- Stephanie M Wolahan
- 1 UCLA Brain Injury Research Center and Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Elliott Lebby
- 1 UCLA Brain Injury Research Center and Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Howard C Mao
- 1 UCLA Brain Injury Research Center and Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - David McArthur
- 1 UCLA Brain Injury Research Center and Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Courtney Real
- 1 UCLA Brain Injury Research Center and Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Paul Vespa
- 1 UCLA Brain Injury Research Center and Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Daniel Braas
- 2 UCLA Metabolomics Center, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Thomas C Glenn
- 1 UCLA Brain Injury Research Center and Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
29
|
Tu TW, Ibrahim WG, Jikaria N, Munasinghe JP, Witko JA, Hammoud DA, Frank JA. On the detection of cerebral metabolic depression in experimental traumatic brain injury using Chemical Exchange Saturation Transfer (CEST)-weighted MRI. Sci Rep 2018; 8:669. [PMID: 29330386 PMCID: PMC5766554 DOI: 10.1038/s41598-017-19094-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 12/21/2017] [Indexed: 12/13/2022] Open
Abstract
Metabolic abnormalities are commonly observed in traumatic brain injury (TBI) patients exhibiting long-term neurological deficits. This study investigated the feasibility and reproducibility of using chemical exchange saturation transfer (CEST) MRI to detect cerebral metabolic depression in experimental TBI. Phantom and in vivo CEST experiments were conducted at 9.4 Tesla to optimize the selective saturation for enhancing the endogenous contrast-weighting of the proton exchanges over the range of glucose proton chemical shifts (glucoCEST) in the resting rat brain. The optimized glucoCEST-weighted imaging was performed on a closed-head model of diffuse TBI in rats with 2-deoxy-D-[14C]-glucose (2DG) autoradiography validation. The results demonstrated that saturation duration of 1‒2 seconds at pulse powers 1.5‒2µT resulted in an improved contrast-to-noise ratio between the gray and white matter comparable to 2DG autoradiographs. The intrasubject (n = 4) and intersubject (n = 3) coefficient of variations for repeated glucoCEST acquisitions (n = 4) ranged between 8‒16%. Optimization for the TBI study revealed that glucoCEST-weighted images with 1.5μT power and 1 s saturation duration revealed the greatest changes in contrast before and after TBI, and positively correlated with 2DG autoradiograph (r = 0.78, p < 0.01, n = 6) observations. These results demonstrate that glucoCEST-weighted imaging may be useful in detecting metabolic abnormalities following TBI.
Collapse
Affiliation(s)
- Tsang-Wei Tu
- Frank Laboratory, Radiology & Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States. .,Center for Neuroscience and Regenerative Medicine, Henry Jackson Foundation, Bethesda, MD, United States. .,Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, United States.
| | - Wael G Ibrahim
- Center for Infectious Disease Imaging, Radiology & Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Neekita Jikaria
- Frank Laboratory, Radiology & Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States.,Center for Neuroscience and Regenerative Medicine, Henry Jackson Foundation, Bethesda, MD, United States.,Acute Stroke Research Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Jeeva P Munasinghe
- Mouse Imaging Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Jaclyn A Witko
- Frank Laboratory, Radiology & Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States.,Center for Neuroscience and Regenerative Medicine, Henry Jackson Foundation, Bethesda, MD, United States
| | - Dima A Hammoud
- Center for Infectious Disease Imaging, Radiology & Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Joseph A Frank
- Frank Laboratory, Radiology & Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States.,National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
30
|
Abstract
Traumatic brain injury is the number one cause of death and disability among the pediatric population in the USA. The heterogeneity of the pediatric population is reflected by both the normal cerebral maturation and the age differences in the causes of TBI, which generate unique age-related pathophysiology responses and recovery profiles. This review will address the normal changes in cerebral glucose metabolism throughout developmental phases and how TBI alters glucose metabolism. Evidence has shown that TBI disrupts the biochemical processing of glucose to energy. This brings to question, "What is the optimal substrate to manage a pediatric TBI patient?" Issues related to glycemic control and alternative substrate metabolism are addressed specifically in regard to pediatric TBI. Research into pediatric glucose metabolism after TBI is limited, and understanding these age-related differences within the pediatric population have great potential to improve support for the injured younger brain.
Collapse
|
31
|
Moreno KX, Harrison CE, Merritt ME, Kovacs Z, Malloy CR, Sherry AD. Hyperpolarized δ-[1- 13 C]gluconolactone as a probe of the pentose phosphate pathway. NMR IN BIOMEDICINE 2017; 30:10.1002/nbm.3713. [PMID: 28272754 PMCID: PMC5502806 DOI: 10.1002/nbm.3713] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/22/2016] [Accepted: 01/23/2017] [Indexed: 05/05/2023]
Abstract
The pentose phosphate pathway (PPP) is thought to be upregulated in trauma (to produce excess NADPH) and in cancer (to provide ribose for nucleotide biosynthesis), but simple methods for detecting changes in flux through this pathway are not available. MRI of hyperpolarized 13 C-enriched metabolites offers considerable potential as a rapid, non-invasive tool for detecting changes in metabolic fluxes. In this study, hyperpolarized δ-[1-13 C]gluconolactone was used as a probe to detect flux through the oxidative portion of the pentose phosphate pathway (PPPox ) in isolated perfused mouse livers. The appearance of hyperpolarized (HP) H13 CO3- within seconds after exposure of livers to HP-δ-[1-13 C]gluconolactone demonstrates that this probe rapidly enters hepatocytes, becomes phosphorylated, and enters the PPPox pathway to produce HP-H13 CO3- after three enzyme catalyzed steps (6P-gluconolactonase, 6-phosphogluconate dehydrogenase, and carbonic anhydrase). Livers perfused with octanoate as their sole energy source show no change in production of H13 CO3- after exposure to low levels of H2 O2 , while livers perfused with glucose and insulin showed a twofold increase in H13 CO3- after exposure to peroxide. This indicates that flux through the PPPox is stimulated by H2 O2 in glucose perfused livers but not in livers perfused with octanoate alone. Subsequent perfusion of livers with non-polarized [1,2-13 C]glucose followed by 1 H NMR analysis of lactate in the perfusate verified that flux through the PPPox is indeed low in healthy livers and modestly higher in peroxide damaged livers. We conclude that hyperpolarized δ-[1-13 C]gluconolactone has the potential to serve as a metabolic imaging probe of this important biological pathway.
Collapse
Affiliation(s)
- Karlos X. Moreno
- Advanced Imaging Research Center, UT Southwestern Medical Center, Dallas, TX 75390
| | - Crystal E. Harrison
- Advanced Imaging Research Center, UT Southwestern Medical Center, Dallas, TX 75390
| | - Matthew E. Merritt
- Advanced Imaging Research Center, UT Southwestern Medical Center, Dallas, TX 75390
- Dept of Radiology, UT Southwestern Medical Center, Dallas, TX 75390
| | - Zoltan Kovacs
- Advanced Imaging Research Center, UT Southwestern Medical Center, Dallas, TX 75390
| | - Craig R. Malloy
- Advanced Imaging Research Center, UT Southwestern Medical Center, Dallas, TX 75390
- Dept of Radiology, UT Southwestern Medical Center, Dallas, TX 75390
- Dept of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390
- VA North Texas Health Care System, Dallas, TX 75216
| | - A. Dean Sherry
- Advanced Imaging Research Center, UT Southwestern Medical Center, Dallas, TX 75390
- Dept of Radiology, UT Southwestern Medical Center, Dallas, TX 75390
- Dept of Chemistry, University of Texas at Dallas, Richardson, TX 75083
- Corresponding Author: A. Dean Sherry; Advanced Imaging Research Center; 5323 Harry Hines Blvd, Dallas, TX 75390; telephone: +1 (214) 645-2730, fax: +1 (214) 645-2744; ; URL: http://www8.utsouthwestern.edu/utsw/home/research/AIRC/index.html
| |
Collapse
|
32
|
Glucose and Intermediary Metabolism and Astrocyte–Neuron Interactions Following Neonatal Hypoxia–Ischemia in Rat. Neurochem Res 2016; 42:115-132. [DOI: 10.1007/s11064-016-2149-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 12/09/2016] [Accepted: 12/10/2016] [Indexed: 11/27/2022]
|
33
|
Van Horn JD, Bhattrai A, Irimia A. Multimodal Imaging of Neurometabolic Pathology due to Traumatic Brain Injury. Trends Neurosci 2016; 40:39-59. [PMID: 27939821 DOI: 10.1016/j.tins.2016.10.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 10/21/2016] [Accepted: 10/25/2016] [Indexed: 12/28/2022]
Abstract
The impact of traumatic brain injury (TBI) involves a combination of complex biochemical processes beginning with the initial insult and lasting for days, months and even years post-trauma. These changes range from neuronal integrity losses to neurotransmitter imbalance and metabolite dysregulation, leading to the release of pro- or anti-apoptotic factors which mediate cell survival or death. Such dynamic processes affecting the brain's neurochemistry can be monitored using a variety of neuroimaging techniques, whose combined use can be particularly useful for understanding patient-specific clinical trajectories. Here, we describe how TBI changes the metabolism of essential neurochemical compounds, summarize how neuroimaging approaches facilitate the study of such alterations, and highlight promising ways in which neuroimaging can be used to investigate post-TBI changes in neurometabolism.
Collapse
Affiliation(s)
- John Darrell Van Horn
- USC Mark and Mary Stevens Neuroimaging and Informatics Institute, 2025 Zonal Avenue, Keck School of Medicine of USC, University of Southern California, Los Angeles, California 90033, USA.
| | - Avnish Bhattrai
- USC Mark and Mary Stevens Neuroimaging and Informatics Institute, 2025 Zonal Avenue, Keck School of Medicine of USC, University of Southern California, Los Angeles, California 90033, USA
| | - Andrei Irimia
- USC Mark and Mary Stevens Neuroimaging and Informatics Institute, 2025 Zonal Avenue, Keck School of Medicine of USC, University of Southern California, Los Angeles, California 90033, USA
| |
Collapse
|
34
|
Shijo K, Sutton RL, Ghavim SS, Harris NG, Bartnik-Olson BL. Metabolic fate of glucose in rats with traumatic brain injury and pyruvate or glucose treatments: A NMR spectroscopy study. Neurochem Int 2016; 102:66-78. [PMID: 27919624 DOI: 10.1016/j.neuint.2016.11.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 11/30/2016] [Accepted: 11/30/2016] [Indexed: 12/27/2022]
Abstract
Administration of sodium pyruvate (SP; 9.08 μmol/kg, i.p.), ethyl pyruvate (EP; 0.34 μmol/kg, i.p.) or glucose (GLC; 11.1 μmol/kg, i.p.) to rats after unilateral controlled cortical impact (CCI) injury has been reported to reduce neuronal loss and improve cerebral metabolism. In the present study these doses of each fuel or 8% saline (SAL; 5.47 nmoles/kg) were administered immediately and at 1, 3, 6 and 23 h post-CCI. At 24 h all CCI groups and non-treated Sham injury controls were infused with [1,2 13C] glucose for 68 min 13C nuclear magnetic resonance (NMR) spectra were obtained from cortex + hippocampus tissues from left (injured) and right (contralateral) hemispheres. All three fuels increased lactate labeling to a similar degree in the injured hemisphere. The amount of lactate labeled via the pentose phosphate and pyruvate recycling (PPP + PR) pathway increased in CCI-SAL and was not improved by SP, EP, and GLC treatments. Oxidative metabolism, as assessed by glutamate labeling, was reduced in CCI-SAL animals. The greatest improvement in oxidative metabolism was observed in animals treated with SP and fewer improvements after EP or GLC treatments. Compared to SAL, all three fuels restored glutamate and glutamine labeling via pyruvate carboxylase (PC), suggesting improved astrocyte metabolism following fuel treatment. Only SP treatments restored the amount of [4 13C] glutamate labeled by the PPP + PR pathway to sham levels. Milder injury effects in the contralateral hemisphere appear normalized by either SP or EP treatments, as increases in the total pool of 13C lactate and labeling of lactate in glycolysis, or decreases in the ratio of PC/PDH labeling of glutamine, were found only for CCI-SAL and CCI-GLC groups compared to Sham. The doses of SP, EP and GLC examined in this study all enhanced lactate labeling and restored astrocyte-specific PC activity but differentially affected neuronal metabolism after CCI injury. The restoration of astrocyte metabolism by all three fuel treatments may partially underlie their abilities to improve cerebral glucose utilization and to reduce neuronal loss following CCI injury.
Collapse
Affiliation(s)
- Katsunori Shijo
- Brain Injury Research Center, Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, Box 956901, CA, USA.
| | - Richard L Sutton
- Brain Injury Research Center, Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, Box 956901, CA, USA.
| | - Sima S Ghavim
- Brain Injury Research Center, Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, Box 956901, CA, USA.
| | - Neil G Harris
- Brain Injury Research Center, Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, Box 956901, CA, USA.
| | | |
Collapse
|
35
|
Wolahan SM, Hirt D, Braas D, Glenn TC. Role of Metabolomics in Traumatic Brain Injury Research. Neurosurg Clin N Am 2016; 27:465-72. [PMID: 27637396 DOI: 10.1016/j.nec.2016.05.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Metabolomics is an important member of the omics community in that it defines which small molecules may be responsible for disease states. This article reviews the essential principles of metabolomics from specimen preparation, chemical analysis, to advanced statistical methods. Metabolomics in traumatic brain injury has so far been underutilized. Future metabolomics-based studies focused on the diagnoses, prognoses, and treatment effects need to be conducted across all types of traumatic brain injury.
Collapse
Affiliation(s)
- Stephanie M Wolahan
- UCLA Brain Injury Research Center, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095, USA; Department of Neurosurgery, David Geffen School of Medicine at UCLA, 300 Stein Plaza, Los Angeles, CA 90095-6901, USA
| | - Daniel Hirt
- UCLA Brain Injury Research Center, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095, USA; Department of Neurosurgery, David Geffen School of Medicine at UCLA, 300 Stein Plaza, Los Angeles, CA 90095-6901, USA
| | - Daniel Braas
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, 570 Westwood Plaza, Los Angeles, CA 90095-1735, USA; UCLA Metabolomics and Proteomics Center, 570 Westwood Plaza, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Thomas C Glenn
- UCLA Brain Injury Research Center, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Los Angeles, CA 90095, USA; Department of Neurosurgery, David Geffen School of Medicine at UCLA, 300 Stein Plaza, Los Angeles, CA 90095-6901, USA.
| |
Collapse
|
36
|
Kilanczyk E, Saraswat Ohri S, Whittemore SR, Hetman M. Antioxidant Protection of NADPH-Depleted Oligodendrocyte Precursor Cells Is Dependent on Supply of Reduced Glutathione. ASN Neuro 2016; 8:8/4/1759091416660404. [PMID: 27449129 PMCID: PMC4962338 DOI: 10.1177/1759091416660404] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 06/29/2016] [Indexed: 01/02/2023] Open
Abstract
The pentose phosphate pathway is the main source of NADPH, which by reducing oxidized glutathione, contributes to antioxidant defenses. Although oxidative stress plays a major role in white matter injury, significance of NADPH for oligodendrocyte survival has not been yet investigated. It is reported here that the NADPH antimetabolite 6-amino-NADP (6AN) was cytotoxic to cultured adult rat spinal cord oligodendrocyte precursor cells (OPCs) as well as OPC-derived oligodendrocytes. The 6AN-induced necrosis was preceded by increased production of superoxide, NADPH depletion, and lower supply of reduced glutathione. Moreover, survival of NADPH-depleted OPCs was improved by the antioxidant drug trolox. Such cells were also protected by physiological concentrations of the neurosteroid dehydroepiandrosterone (10−8 M). The protection by dehydroepiandrosterone was associated with restoration of reduced glutathione, but not NADPH, and was sensitive to inhibition of glutathione synthesis. A similar protective mechanism was engaged by the cAMP activator forskolin or the G protein-coupled estrogen receptor (GPER/GPR30) ligand G1. Finally, treatment with the glutathione precursor N-acetyl cysteine reduced cytotoxicity of 6AN. Taken together, NADPH is critical for survival of OPCs by supporting their antioxidant defenses. Consequently, injury-associated inhibition of the pentose phosphate pathway may be detrimental for the myelination or remyelination potential of the white matter. Conversely, steroid hormones and cAMP activators may promote survival of NADPH-deprived OPCs by increasing a NADPH-independent supply of reduced glutathione. Therefore, maintenance of glutathione homeostasis appears as a critical effector mechanism for OPC protection against NADPH depletion and preservation of the regenerative potential of the injured white matter.
Collapse
Affiliation(s)
- Ewa Kilanczyk
- Kentucky Spinal Cord Injury Research Center, University of Louisville, KY, USA Department of Neurological Surgery, University of Louisville, KY, USA
| | - Sujata Saraswat Ohri
- Kentucky Spinal Cord Injury Research Center, University of Louisville, KY, USA Department of Neurological Surgery, University of Louisville, KY, USA
| | - Scott R Whittemore
- Kentucky Spinal Cord Injury Research Center, University of Louisville, KY, USA Department of Neurological Surgery, University of Louisville, KY, USA Department of Anatomical Sciences and Neurobiology, University of Louisville, KY, USA
| | - Michal Hetman
- Kentucky Spinal Cord Injury Research Center, University of Louisville, KY, USA Department of Neurological Surgery, University of Louisville, KY, USA Department of Pharmacology and Toxicology, University of Louisville, KY, USA
| |
Collapse
|
37
|
Moro N, Ghavim SS, Harris NG, Hovda DA, Sutton RL. Pyruvate treatment attenuates cerebral metabolic depression and neuronal loss after experimental traumatic brain injury. Brain Res 2016; 1642:270-277. [PMID: 27059390 DOI: 10.1016/j.brainres.2016.04.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 04/01/2016] [Accepted: 04/04/2016] [Indexed: 12/20/2022]
Abstract
Experimental traumatic brain injury (TBI) is known to produce an acute increase in cerebral glucose utilization, followed rapidly by a generalized cerebral metabolic depression. The current studies determined effects of single or multiple treatments with sodium pyruvate (SP; 1000mg/kg, i.p.) or ethyl pyruvate (EP; 40mg/kg, i.p.) on cerebral glucose metabolism and neuronal injury in rats with unilateral controlled cortical impact (CCI) injury. In Experiment 1 a single treatment was given immediately after CCI. SP significantly improved glucose metabolism in 3 of 13 brain regions while EP improved metabolism in 7 regions compared to saline-treated controls at 24h post-injury. Both SP and EP produced equivalent and significant reductions in dead/dying neurons in cortex and hippocampus at 24h post-CCI. In Experiment 2 SP or EP were administered immediately (time 0) and at 1, 3 and 6h post-CCI. Multiple SP treatments also significantly attenuated TBI-induced reductions in cerebral glucose metabolism (in 4 brain regions) 24h post-CCI, as did multiple injections of EP (in 4 regions). The four pyruvate treatments produced significant neuroprotection in cortex and hippocampus 1day after CCI, similar to that found with a single SP or EP treatment. Thus, early administration of pyruvate compounds enhanced cerebral glucose metabolism and neuronal survival, with 40mg/kg of EP being as effective as 1000mg/kg of SP, and multiple treatments within 6h of injury did not improve upon outcomes seen following a single treatment.
Collapse
Affiliation(s)
- Nobuhiro Moro
- UCLA Brain Injury Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-6901, USA; Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-6901, USA.
| | - Sima S Ghavim
- UCLA Brain Injury Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-6901, USA; Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-6901, USA.
| | - Neil G Harris
- UCLA Brain Injury Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-6901, USA; Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-6901, USA.
| | - David A Hovda
- UCLA Brain Injury Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-6901, USA; Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-6901, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-6901, USA.
| | - Richard L Sutton
- UCLA Brain Injury Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-6901, USA; Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-6901, USA.
| |
Collapse
|
38
|
de Lima Oliveira M, Bor-Seng-Shu E, Simm RF, Vilas Boas T, Pires Aguiar PH. Commentary: Systemic, Local, and Imaging Biomarkers of Brain Injury: More Needed, and Better Use of Those Already Established? Front Neurol 2016; 7:34. [PMID: 27047442 PMCID: PMC4800171 DOI: 10.3389/fneur.2016.00034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 02/29/2016] [Indexed: 11/18/2022] Open
Affiliation(s)
- Marcelo de Lima Oliveira
- Neurology Department, Hospital Santa Paula, São Paulo, Brazil
- *Correspondence: Marcelo de Lima Oliveira,
| | | | | | | | | |
Collapse
|
39
|
Iglesias J, Morales L, Barreto GE. Metabolic and Inflammatory Adaptation of Reactive Astrocytes: Role of PPARs. Mol Neurobiol 2016; 54:2518-2538. [PMID: 26984740 DOI: 10.1007/s12035-016-9833-2] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/04/2016] [Indexed: 01/10/2023]
Abstract
Astrocyte-mediated inflammation is associated with degenerative pathologies such as Alzheimer's and Parkinson's diseases and multiple sclerosis. The acute inflammation and morphological and metabolic changes that astrocytes develop after the insult are known as reactive astroglia or astrogliosis that is an important response to protect and repair the lesion. Astrocytes optimize their metabolism to produce lactate, glutamate, and ketone bodies in order to provide energy to the neurons that are deprived of nutrients upon insult. Firstly, we review the basis of inflammation and morphological changes of the different cell population implicated in reactive gliosis. Next, we discuss the more active metabolic pathways in healthy astrocytes and explain the metabolic response of astrocytes to the insult in different pathologies and which metabolic alterations generate complications in these diseases. We emphasize the role of peroxisome proliferator-activated receptors isotypes in the inflammatory and metabolic adaptation of astrogliosis developed in ischemia or neurodegenerative diseases. Based on results reported in astrocytes and other cells, we resume and hypothesize the effect of peroxisome proliferator-activated receptor (PPAR) activation with ligands on different metabolic pathways in order to supply energy to the neurons. The activation of selective PPAR isotype activity may serve as an input to better understand the role played by these receptors on the metabolic and inflammatory compensation of astrogliosis and might represent an opportunity to develop new therapeutic strategies against traumatic brain injuries and neurodegenerative diseases.
Collapse
Affiliation(s)
- José Iglesias
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, DC, Colombia.
| | - Ludis Morales
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, DC, Colombia
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, DC, Colombia
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
- Universidad Científica del Sur, Lima, Peru
| |
Collapse
|
40
|
Amorini AM, Lazzarino G, Di Pietro V, Signoretti S, Lazzarino G, Belli A, Tavazzi B. Metabolic, enzymatic and gene involvement in cerebral glucose dysmetabolism after traumatic brain injury. Biochim Biophys Acta Mol Basis Dis 2016; 1862:679-687. [PMID: 26844378 DOI: 10.1016/j.bbadis.2016.01.023] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 01/29/2016] [Indexed: 12/16/2022]
Abstract
In this study, the metabolic, enzymatic and gene changes causing cerebral glucose dysmetabolism following graded diffuse traumatic brain injury (TBI) were evaluated. TBI was induced in rats by dropping 450g from 1 (mild TBI; mTBI) or 2m height (severe TBI; sTBI). After 6, 12, 24, 48, and 120h gene expressions and enzymatic activities of glycolysis and pentose phosphate pathway (PPP) enzymes, and levels of lactate, ATP, ADP, ATP/ADP (indexing mitochondrial phosphorylating capacity), NADP(+), NADPH and GSH were determined in whole brain extracts (n=9 rats at each time for both TBI levels). Sham-operated animals (n=9) were used as controls. Results demonstrated that mTBI caused a late increase (48-120h post injury) of glycolytic gene expression and enzymatic activities, concomitantly with mitochondrial functional recovery (ATP and ATP/ADP normalization). No changes in lactate and PPP genes and enzymes, were accompanied by transient decrease in GSH, NADP(+), NADPH and NADPH/NADP(+). Animals following sTBI showed early increase (6-24h post injury) of glycolytic gene expression and enzymatic activities, occurring during mitochondrial malfunctioning (50% decrease in ATP and ATP/ADP). Higher lactate and lower GSH, NADP(+), NADPH, NADPH/NADP(+) than controls were recorded at anytime post injury (p<0.01). Both TBI levels caused metabolic and gene changes affecting glucose metabolism. Following mTBI, increased glucose flux through glycolysis is coupled to mitochondrial glucose oxidation. "True" hyperglycolysis occurs only after sTBI, where metabolic changes, caused by depressed mitochondrial phosphorylating capacity, act on genes causing net glycolytic flux increase uncoupled from mitochondrial glucose oxidation.
Collapse
Affiliation(s)
- Angela Maria Amorini
- Institute of Biochemistry and Clinical Biochemistry, Catholic University of Rome, Largo F. Vito 1, 00168 Rome, Italy.
| | - Giacomo Lazzarino
- Institute of Biochemistry and Clinical Biochemistry, Catholic University of Rome, Largo F. Vito 1, 00168 Rome, Italy.
| | - Valentina Di Pietro
- Neurobiology, School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, B15 2TT Birmingham, UK.
| | - Stefano Signoretti
- Division of Neurosurgery, Department of Neurosciences Head and Neck Surgery, S. Camillo Hospital, Circonvallazione Gianicolense 87, 00152 Rome, Italy.
| | - Giuseppe Lazzarino
- Department of Biomedical and Biotechnological Sciences, Division of Medical Biochemistry, University of Catania, Viale A. Doria 6, 95125 Catania, Italy.
| | - Antonio Belli
- Neurobiology, School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, B15 2TT Birmingham, UK; National Institute for Health Research Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital, Edgbaston, B15 2TH Birmingham, UK.
| | - Barbara Tavazzi
- Institute of Biochemistry and Clinical Biochemistry, Catholic University of Rome, Largo F. Vito 1, 00168 Rome, Italy.
| |
Collapse
|
41
|
Harada S, Matsuura W, Takano M, Tokuyama S. Withdrawal: Proteomic Profiling in the Spinal Cord and Sciatic Nerve in a Global Cerebral Ischemia-Induced Mechanical Allodynia Mouse Model. Biol Pharm Bull 2016; 39:230-8. [DOI: 10.1248/bpb.b15-00647] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Shinichi Harada
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University
| | - Wataru Matsuura
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University
| | - Masaoki Takano
- Department of Life Sciences Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University
| | - Shogo Tokuyama
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University
| |
Collapse
|
42
|
Jayakumar AR, Bak LK, Rama Rao KV, Waagepetersen HS, Schousboe A, Norenberg MD. Neuronal Cell Death Induced by Mechanical Percussion Trauma in Cultured Neurons is not Preceded by Alterations in Glucose, Lactate and Glutamine Metabolism. Neurochem Res 2016; 41:307-15. [PMID: 26729365 DOI: 10.1007/s11064-015-1801-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/07/2015] [Accepted: 12/09/2015] [Indexed: 11/24/2022]
Abstract
Traumatic brain injury (TBI) is a devastating neurological disorder that usually presents in acute and chronic forms. Brain edema and associated increased intracranial pressure in the early phase following TBI are major consequences of acute trauma. On the other hand, neuronal injury, leading to neurobehavioral and cognitive impairments, that usually develop months to years after single or repetitive episodes of head trauma, are major consequences of chronic TBI. The molecular mechanisms responsible for TBI-induced injury, however, are unclear. Recent studies have suggested that early mitochondrial dysfunction and subsequent energy failure play a role in the pathogenesis of TBI. We therefore examined whether oxidative metabolism of (13)C-labeled glucose, lactate or glutamine is altered early following in vitro mechanical percussion-induced trauma (5 atm) to neurons (4-24 h), and whether such events contribute to the development of neuronal injury. Cell viability was assayed using the release of the cytoplasmic enzyme lactate dehydrogenase (LDH), together with fluorescence-based cell staining (calcein and ethidium homodimer-1 for live and dead cells, respectively). Trauma had no effect on the LDH release in neurons from 1 to 18 h. However, a significant increase in LDH release was detected at 24 h after trauma. Similar findings were identified when traumatized neurons were stained with fluorescent markers. Additionally (13)C-labeling of glutamate showed a small, but statistically significant decrease at 14 h after trauma. However, trauma had no effect on the cycling ratio of the TCA cycle at any time-period examined. These findings indicate that trauma does not cause a disturbance in oxidative metabolism of any of the substrates used for neurons. Accordingly, such metabolic disturbance does not appear to contribute to the neuronal death in the early stages following trauma.
Collapse
Affiliation(s)
- A R Jayakumar
- Laboratory of Neuropathology, Veterans Affairs Medical Center, Miami, FL, USA
| | - L K Bak
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100, Copenhagen, Denmark
| | - K V Rama Rao
- Laboratory of Neuropathology, Veterans Affairs Medical Center, Miami, FL, USA
| | - H S Waagepetersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100, Copenhagen, Denmark
| | - A Schousboe
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100, Copenhagen, Denmark
| | - M D Norenberg
- Laboratory of Neuropathology, Veterans Affairs Medical Center, Miami, FL, USA. .,Department of Pathology (D-33), University of Miami School of Medicine, P.O. Box 016960, Miami, FL, 33101, USA. .,Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, Miami, FL, USA.
| |
Collapse
|
43
|
Prins ML, Matsumoto J. Metabolic Response of Pediatric Traumatic Brain Injury. J Child Neurol 2016; 31:28-34. [PMID: 25336427 PMCID: PMC4405388 DOI: 10.1177/0883073814549244] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 07/21/2014] [Indexed: 12/26/2022]
Abstract
Traumatic brain injury (TBI) in the pediatric brain presents unique challenges as the complex cascades of metabolic and biochemical responses to TBI are further complicated ongoing maturational changes of the developing brain. TBIs of all severities have been shown to significantly alter metabolism and hormones which impair the ability of the brain to process glucose for cellular energy. Under these conditions, the brain's primary fuel (glucose) becomes a less favorable fuel and the ability of the younger brain to revert to ketone metabolism can an advantage. This review addresses the potential of alternative substrate metabolic intervention as a logical pediatric TBI neuroprotective strategy.
Collapse
Affiliation(s)
- Mayumi L Prins
- Department of Neurosurgery, University of California Los Angeles, Los Angeles, CA, USA Brain Injury Research Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Joyce Matsumoto
- Department of Pediatrics, Division of Pediatric Neurology, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
44
|
McKenna MC, Scafidi S, Robertson CL. Metabolic Alterations in Developing Brain After Injury: Knowns and Unknowns. Neurochem Res 2015; 40:2527-43. [PMID: 26148530 PMCID: PMC4961252 DOI: 10.1007/s11064-015-1600-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 04/10/2015] [Accepted: 05/02/2015] [Indexed: 12/21/2022]
Abstract
Brain development is a highly orchestrated complex process. The developing brain utilizes many substrates including glucose, ketone bodies, lactate, fatty acids and amino acids for energy, cell division and the biosynthesis of nucleotides, proteins and lipids. Metabolism is crucial to provide energy for all cellular processes required for brain development and function including ATP formation, synaptogenesis, synthesis, release and uptake of neurotransmitters, maintaining ionic gradients and redox status, and myelination. The rapidly growing population of infants and children with neurodevelopmental and cognitive impairments and life-long disability resulting from developmental brain injury is a significant public health concern. Brain injury in infants and children can have devastating effects because the injury is superimposed on the high metabolic demands of the developing brain. Acute injury in the pediatric brain can derail, halt or lead to dysregulation of the complex and highly regulated normal developmental processes. This paper provides a brief review of metabolism in developing brain and alterations found clinically and in animal models of developmental brain injury. The metabolic changes observed in three major categories of injury that can result in life-long cognitive and neurological disabilities, including neonatal hypoxia-ischemia, pediatric traumatic brain injury, and brain injury secondary to prematurity are reviewed.
Collapse
Affiliation(s)
- Mary C McKenna
- Department of Pediatrics and Program in Neuroscience, University of Maryland School of Medicine, 655 W. Baltimore St., Room 13-019, Baltimore, MD, 21201, USA.
| | - Susanna Scafidi
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Courtney L Robertson
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
45
|
McKenna MC, Scafidi S, Robertson CL. Metabolic Alterations in Developing Brain After Injury: Knowns and Unknowns. Neurochem Res 2015. [PMID: 26148530 DOI: 10.1007/s11064‐015‐1600‐7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Brain development is a highly orchestrated complex process. The developing brain utilizes many substrates including glucose, ketone bodies, lactate, fatty acids and amino acids for energy, cell division and the biosynthesis of nucleotides, proteins and lipids. Metabolism is crucial to provide energy for all cellular processes required for brain development and function including ATP formation, synaptogenesis, synthesis, release and uptake of neurotransmitters, maintaining ionic gradients and redox status, and myelination. The rapidly growing population of infants and children with neurodevelopmental and cognitive impairments and life-long disability resulting from developmental brain injury is a significant public health concern. Brain injury in infants and children can have devastating effects because the injury is superimposed on the high metabolic demands of the developing brain. Acute injury in the pediatric brain can derail, halt or lead to dysregulation of the complex and highly regulated normal developmental processes. This paper provides a brief review of metabolism in developing brain and alterations found clinically and in animal models of developmental brain injury. The metabolic changes observed in three major categories of injury that can result in life-long cognitive and neurological disabilities, including neonatal hypoxia-ischemia, pediatric traumatic brain injury, and brain injury secondary to prematurity are reviewed.
Collapse
Affiliation(s)
- Mary C McKenna
- Department of Pediatrics and Program in Neuroscience, University of Maryland School of Medicine, 655 W. Baltimore St., Room 13-019, Baltimore, MD, 21201, USA.
| | - Susanna Scafidi
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Courtney L Robertson
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
46
|
Jalloh I, Carpenter KLH, Helmy A, Carpenter TA, Menon DK, Hutchinson PJ. Glucose metabolism following human traumatic brain injury: methods of assessment and pathophysiological findings. Metab Brain Dis 2015; 30:615-32. [PMID: 25413449 PMCID: PMC4555200 DOI: 10.1007/s11011-014-9628-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Accepted: 11/03/2014] [Indexed: 02/02/2023]
Abstract
The pathophysiology of traumatic brain (TBI) injury involves changes to glucose uptake into the brain and its subsequent metabolism. We review the methods used to study cerebral glucose metabolism with a focus on those used in clinical TBI studies. Arterio-venous measurements provide a global measure of glucose uptake into the brain. Microdialysis allows the in vivo sampling of brain extracellular fluid and is well suited to the longitudinal assessment of metabolism after TBI in the clinical setting. A recent novel development is the use of microdialysis to deliver glucose and other energy substrates labelled with carbon-13, which allows the metabolism of glucose and other substrates to be tracked. Positron emission tomography and magnetic resonance spectroscopy allow regional differences in metabolism to be assessed. We summarise the data published from these techniques and review their potential uses in the clinical setting.
Collapse
Affiliation(s)
- Ibrahim Jalloh
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Box 167 Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK,
| | | | | | | | | | | |
Collapse
|
47
|
Glucose administration after traumatic brain injury exerts some benefits and no adverse effects on behavioral and histological outcomes. Brain Res 2015; 1614:94-104. [PMID: 25911580 DOI: 10.1016/j.brainres.2015.04.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 04/10/2015] [Accepted: 04/11/2015] [Indexed: 11/22/2022]
Abstract
The impact of hyperglycemia after traumatic brain injury (TBI), and even the administration of glucose-containing solutions to head injured patients, remains controversial. In the current study adult male Sprague-Dawley rats were tested on behavioral tasks and then underwent surgery to induce sham injury or unilateral controlled cortical impact (CCI) injury followed by injections (i.p.) with either a 50% glucose solution (Glc; 2g/kg) or an equivalent volume of either 0.9% or 8% saline (Sal) at 0, 1, 3 and 6h post-injury. The type of saline treatment did not significantly affect any outcome measures, so these data were combined. Rats with CCI had significant deficits in beam-walking traversal time and rating scores (p's < 0.001 versus sham) that recovered over test sessions from 1 to 13 days post-injury (p's < 0.001), but these beam-walking deficits were not affected by Glc versus Sal treatments. Persistent post-CCI deficits in forelimb contraflexion scores and forelimb tactile placing ability were also not differentially affected by Glc or Sal treatments. However, deficits in latency to retract the right hind limb after limb extension were significantly attenuated in the CCI-Glc group (p < 0.05 versus CCI-Sal). Both CCI groups were significantly impaired in a plus maze test of spatial working memory on days 4, 9 and 14 post-surgery (p < 0.001 versus sham), and there was no effect of Glc versus Sal on this cognitive outcome measure. At 15 days post-surgery the loss of cortical tissue volume (p < 0.001 versus sham) was significantly less in the CCI-Glc group (30.0%; p < 0.05) compared to the CCI-Sal group (35.7%). Counts of surviving hippocampal hilar neurons revealed a significant (~40%) loss ipsilateral to CCI (p < 0.001 versus sham), but neuronal loss in the hippocampus was not different in the CCI-Sal and CCI-Glc groups. Taken together, these results indicate that an early elevation of blood glucose may improve some neurological outcomes and, importantly, the induction of hyperglycemia after isolated TBI did not adversely affect any sensorimotor, cognitive or histological outcomes.
Collapse
|
48
|
Carpenter KLH, Jalloh I, Hutchinson PJ. Glycolysis and the significance of lactate in traumatic brain injury. Front Neurosci 2015; 9:112. [PMID: 25904838 PMCID: PMC4389375 DOI: 10.3389/fnins.2015.00112] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 03/16/2015] [Indexed: 01/19/2023] Open
Abstract
In traumatic brain injury (TBI) patients, elevation of the brain extracellular lactate concentration and the lactate/pyruvate ratio are well-recognized, and are associated statistically with unfavorable clinical outcome. Brain extracellular lactate was conventionally regarded as a waste product of glucose, when glucose is metabolized via glycolysis (Embden-Meyerhof-Parnas pathway) to pyruvate, followed by conversion to lactate by the action of lactate dehydrogenase, and export of lactate into the extracellular fluid. In TBI, glycolytic lactate is ascribed to hypoxia or mitochondrial dysfunction, although the precise nature of the latter is incompletely understood. Seemingly in contrast to lactate's association with unfavorable outcome is a growing body of evidence that lactate can be beneficial. The idea that the brain can utilize lactate by feeding into the tricarboxylic acid (TCA) cycle of neurons, first published two decades ago, has become known as the astrocyte-neuron lactate shuttle hypothesis. Direct evidence of brain utilization of lactate was first obtained 5 years ago in a cerebral microdialysis study in TBI patients, where administration of (13)C-labeled lactate via the microdialysis catheter and simultaneous collection of the emerging microdialysates, with (13)C NMR analysis, revealed (13)C labeling in glutamine consistent with lactate utilization via the TCA cycle. This suggests that where neurons are too damaged to utilize the lactate produced from glucose by astrocytes, i.e., uncoupling of neuronal and glial metabolism, high extracellular levels of lactate would accumulate, explaining the association between high lactate and poor outcome. Recently, an intravenous exogenous lactate supplementation study in TBI patients revealed evidence for a beneficial effect judged by surrogate endpoints. Here we review the current state of knowledge about glycolysis and lactate in TBI, how it can be measured in patients, and whether it can be modulated to achieve better clinical outcome.
Collapse
Affiliation(s)
- Keri L H Carpenter
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge Cambridge, UK ; Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge Cambridge, UK
| | - Ibrahim Jalloh
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge Cambridge, UK
| | - Peter J Hutchinson
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge Cambridge, UK ; Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge Cambridge, UK
| |
Collapse
|
49
|
de Lima Oliveira M, Kairalla AC, Fonoff ET, Martinez RCR, Teixeira MJ, Bor-Seng-Shu E. Cerebral microdialysis in traumatic brain injury and subarachnoid hemorrhage: state of the art. Neurocrit Care 2015; 21:152-62. [PMID: 24072457 DOI: 10.1007/s12028-013-9884-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cerebral microdialysis (CMD) is a laboratory tool that provides on-line analysis of brain biochemistry via a thin, fenestrated, double-lumen dialysis catheter that is inserted into the interstitium of the brain. A solute is slowly infused into the catheter at a constant velocity. The fenestrated membranes at the tip of the catheter permit free diffusion of molecules between the brain interstitium and the perfusate, which is subsequently collected for laboratory analysis. The major molecules studied using this method are glucose, lactate, pyruvate, glutamate, and glycerol. The collected substances provide insight into the neurochemical features of secondary injury following traumatic brain injury (TBI) and subarachnoid hemorrhage (SAH) and valuable information about changes in brain metabolism within a short time frame. In this review, the authors detail the CMD technique and its associated markers and then describe pertinent findings from the literature about the clinical application of CMD in TBI and SAH.
Collapse
Affiliation(s)
- Marcelo de Lima Oliveira
- Division of Neurological Surgery, Hospital das Clinicas, School of Medicine, University of São Paulo, Rua Loefgreen, 1.272 - Vila Clementino, São Paulo, SP, 04040-001, Brazil
| | | | | | | | | | | |
Collapse
|
50
|
Brekke E, Morken TS, Sonnewald U. Glucose metabolism and astrocyte-neuron interactions in the neonatal brain. Neurochem Int 2015; 82:33-41. [PMID: 25684072 DOI: 10.1016/j.neuint.2015.02.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 02/07/2015] [Accepted: 02/09/2015] [Indexed: 10/24/2022]
Abstract
Glucose is essentially the sole fuel for the adult brain and the mapping of its metabolism has been extensive in the adult but not in the neonatal brain, which is believed to rely mainly on ketone bodies for energy supply. However, glucose is absolutely indispensable for normal development and recent studies have shed light on glycolysis, the pentose phosphate pathway and metabolic interactions between astrocytes and neurons in the 7-day-old rat brain. Appropriately (13)C labeled glucose was used to distinguish between glycolysis and the pentose phosphate pathway during development. Experiments using (13)C labeled acetate provided insight into the GABA-glutamate-glutamine cycle between astrocytes and neurons. It could be shown that in the neonatal brain the part of this cycle that transfers glutamine from astrocytes to neurons is operating efficiently while, in contrast, little glutamate is shuttled from neurons to astrocytes. This lack of glutamate for glutamine synthesis is compensated for by anaplerosis via increased pyruvate carboxylation relative to that in the adult brain. Furthermore, compared to adults, relatively more glucose is prioritized to the pentose phosphate pathway than glycolysis and pyruvate dehydrogenase activity. The reported developmental differences in glucose metabolism and neurotransmitter synthesis may determine the ability of the brain at various ages to resist excitotoxic insults such as hypoxia-ischemia.
Collapse
Affiliation(s)
- Eva Brekke
- Department of Neuroscience, Norwegian University of Science and Technology, Trondheim N-7489, Norway; Department of Pediatrics, Division of Pediatrics, Obstetrics and Women's Health, Nordland Hospital Trust, Bodo, Norway
| | - Tora Sund Morken
- Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology (NTNU), Trondheim N-7489, Norway; Department of Ophthalmology, St. Olav's Hospital HF, Trondheim, Norway
| | - Ursula Sonnewald
- Department of Neuroscience, Norwegian University of Science and Technology, Trondheim N-7489, Norway.
| |
Collapse
|