1
|
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] [Academic Contribution 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
|
2
|
Nisar S, Haris M. Neuroimaging genetics approaches to identify new biomarkers for the early diagnosis of autism spectrum disorder. Mol Psychiatry 2023; 28:4995-5008. [PMID: 37069342 PMCID: PMC11041805 DOI: 10.1038/s41380-023-02060-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 08/01/2022] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 04/19/2023]
Abstract
Autism-spectrum disorders (ASDs) are developmental disabilities that manifest in early childhood and are characterized by qualitative abnormalities in social behaviors, communication skills, and restrictive or repetitive behaviors. To explore the neurobiological mechanisms in ASD, extensive research has been done to identify potential diagnostic biomarkers through a neuroimaging genetics approach. Neuroimaging genetics helps to identify ASD-risk genes that contribute to structural and functional variations in brain circuitry and validate biological changes by elucidating the mechanisms and pathways that confer genetic risk. Integrating artificial intelligence models with neuroimaging data lays the groundwork for accurate diagnosis and facilitates the identification of early diagnostic biomarkers for ASD. This review discusses the significance of neuroimaging genetics approaches to gaining a better understanding of the perturbed neurochemical system and molecular pathways in ASD and how these approaches can detect structural, functional, and metabolic changes and lead to the discovery of novel biomarkers for the early diagnosis of ASD.
Collapse
Affiliation(s)
- Sabah Nisar
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, Doha, Qatar
- Department of Diagnostic Imaging, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Mohammad Haris
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, Doha, Qatar.
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Laboratory Animal Research Center, Qatar University, Doha, Qatar.
| |
Collapse
|
3
|
Vatte S, Ugale R. HIF-1, an important regulator in potential new therapeutic approaches to ischemic stroke. Neurochem Int 2023; 170:105605. [PMID: 37657765 DOI: 10.1016/j.neuint.2023.105605] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/24/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/03/2023]
Abstract
Ischemic stroke is a leading cause of disability and mortality worldwide due to the narrow therapeutic window of the only approved therapies like intravenous thrombolysis and thrombectomy. Hypoxia inducible factor-1α (HIF-1α) is a sensitive regulator of oxygen homeostasis, and its expression is rapidly induced after hypoxia/ischemia. It plays an extensive role in the pathophysiology of stroke by regulating multiple pathways including glucose metabolism, angiogenesis, neuronal survival, neuroinflammation and blood brain barrier regulation. Here, we give a brief overview of the HIF-1α-targeting strategies currently under investigation and summarise recent research on how HIF-1α is regulated in various brain cells, including neurons and microglia, at various stages in ischemic stroke. The roles of HIF-1 in stroke varies with ischemic time and degree of ischemia, are still up for debate. More focus has been placed on prospective HIF-1α targeting drugs, such as HIF-1α activator, HIF-1α stabilizers, and natural compounds. In this review, we have highlighted the regulation of HIF-1α in the novel therapeutic approaches for treatment of stroke.
Collapse
Affiliation(s)
- Sneha Vatte
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, 440033, India.
| | - Rajesh Ugale
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, 440033, India.
| |
Collapse
|
4
|
Villapol S, Janatpour ZC, Affram KO, Symes AJ. The Renin Angiotensin System as a Therapeutic Target in Traumatic Brain Injury. Neurotherapeutics 2023; 20:1565-1591. [PMID: 37759139 PMCID: PMC10684482 DOI: 10.1007/s13311-023-01435-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Traumatic brain injury (TBI) is a major public health problem, with limited pharmacological options available beyond symptomatic relief. The renin angiotensin system (RAS) is primarily known as a systemic endocrine regulatory system, with major roles controlling blood pressure and fluid homeostasis. Drugs that target the RAS are used to treat hypertension, heart failure and kidney disorders. They have now been used chronically by millions of people and have a favorable safety profile. In addition to the systemic RAS, it is now appreciated that many different organ systems, including the brain, have their own local RAS. The major ligand of the classic RAS, Angiotensin II (Ang II) acts predominantly through the Ang II Type 1 receptor (AT1R), leading to vasoconstriction, inflammation, and heightened oxidative stress. These processes can exacerbate brain injuries. Ang II receptor blockers (ARBs) are AT1R antagonists. They have been shown in several preclinical studies to enhance recovery from TBI in rodents through improvements in molecular, cellular and behavioral correlates of injury. ARBs are now under consideration for clinical trials in TBI. Several different RAS peptides that signal through receptors distinct from the AT1R, are also potential therapeutic targets for TBI. The counter regulatory RAS pathway has actions that oppose those stimulated by AT1R signaling. This alternative pathway has many beneficial effects on cells in the central nervous system, bringing about vasodilation, and having anti-inflammatory and anti-oxidative stress actions. Stimulation of this pathway also has potential therapeutic value for the treatment of TBI. This comprehensive review will provide an overview of the various components of the RAS, with a focus on their direct relevance to TBI pathology. It will explore different therapeutic agents that modulate this system and assess their potential efficacy in treating TBI patients.
Collapse
Affiliation(s)
- Sonia Villapol
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, USA
| | - Zachary C Janatpour
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Kwame O Affram
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Aviva J Symes
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
5
|
Kathote G, Ma Q, Angulo G, Chen H, Jakkamsetti V, Dobariya A, Good LB, Posner B, Park JY, Pascual JM. Identification of Glucose Transport Modulators In Vitro and Method for Their Deep Learning Neural Network Behavioral Evaluation in Glucose Transporter 1-Deficient Mice. J Pharmacol Exp Ther 2023; 384:393-405. [PMID: 36635085 DOI: 10.1124/jpet.122.001428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/26/2022] [Revised: 12/07/2022] [Accepted: 12/27/2022] [Indexed: 01/14/2023] Open
Abstract
Metabolic flux augmentation via glucose transport activation may be desirable in glucose transporter 1 (Glut1) deficiency syndrome (G1D) and dementia, whereas suppression might prove useful in cancer. Using lung adenocarcinoma cells that predominantly express Glut1 relative to other glucose transporters, we screened 9,646 compounds for effects on the accumulation of an extracellularly applied fluorescent glucose analog. Five drugs currently prescribed for unrelated indications or preclinically characterized robustly enhanced intracellular fluorescence. Additionally identified were 37 novel activating and nine inhibitory compounds lacking previous biologic characterization. Because few glucose-related mechanistic or pharmacological studies were available for these compounds, we developed a method to quantify G1D mouse behavior to infer potential therapeutic value. To this end, we designed a five-track apparatus to record and evaluate spontaneous locomotion videos. We applied this to a G1D mouse model that replicates the ataxia and other manifestations cardinal to the human disorder. Because the first two drugs that we examined in this manner (baclofen and acetazolamide) exerted various impacts on several gait aspects, we used deep learning neural networks to more comprehensively assess drug effects. Using this method, 49 locomotor parameters differentiated G1D from control mice. Thus, we used parameter modifiability to quantify efficacy on gait. We tested this by measuring the effects of saline as control and glucose as G1D therapy. The results indicate that this in vivo approach can estimate preclinical suitability from the perspective of G1D locomotion. This justifies the use of this method to evaluate our drugs or other interventions and sort candidates for further investigation. SIGNIFICANCE STATEMENT: There are few or no activators and few clinical inhibitors of glucose transport. Using Glut1-rich cells exposed to a glucose analog, we identified, in highthroughput fashion, a series of novel modulators. Some were drugs used to modify unrelated processes and some represented large but little studied chemical compound families. To facilitate their preclinical efficacy characterization regardless of potential mechanism of action, we developed a gait testing platform for deep learning neural network analysis of drug impact on Glut1-deficient mouse locomotion.
Collapse
Affiliation(s)
- Gauri Kathote
- Rare Brain Disorders Program, Department of Neurology (G.K., Q.M., G.A., V.J., A.D., L.B.G., J.M.P.), Department of Biochemistry (H.C., B.P.), Department of Pathology (J.Y.P.), Department of Physiology (J.M.P.), Department of Pediatrics (J.M.P.), and Eugene McDermott Center for Human Growth & Development/Center for Human Genetics (J.Y.P., J.M.P.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Qian Ma
- Rare Brain Disorders Program, Department of Neurology (G.K., Q.M., G.A., V.J., A.D., L.B.G., J.M.P.), Department of Biochemistry (H.C., B.P.), Department of Pathology (J.Y.P.), Department of Physiology (J.M.P.), Department of Pediatrics (J.M.P.), and Eugene McDermott Center for Human Growth & Development/Center for Human Genetics (J.Y.P., J.M.P.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Gustavo Angulo
- Rare Brain Disorders Program, Department of Neurology (G.K., Q.M., G.A., V.J., A.D., L.B.G., J.M.P.), Department of Biochemistry (H.C., B.P.), Department of Pathology (J.Y.P.), Department of Physiology (J.M.P.), Department of Pediatrics (J.M.P.), and Eugene McDermott Center for Human Growth & Development/Center for Human Genetics (J.Y.P., J.M.P.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Hong Chen
- Rare Brain Disorders Program, Department of Neurology (G.K., Q.M., G.A., V.J., A.D., L.B.G., J.M.P.), Department of Biochemistry (H.C., B.P.), Department of Pathology (J.Y.P.), Department of Physiology (J.M.P.), Department of Pediatrics (J.M.P.), and Eugene McDermott Center for Human Growth & Development/Center for Human Genetics (J.Y.P., J.M.P.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Vikram Jakkamsetti
- Rare Brain Disorders Program, Department of Neurology (G.K., Q.M., G.A., V.J., A.D., L.B.G., J.M.P.), Department of Biochemistry (H.C., B.P.), Department of Pathology (J.Y.P.), Department of Physiology (J.M.P.), Department of Pediatrics (J.M.P.), and Eugene McDermott Center for Human Growth & Development/Center for Human Genetics (J.Y.P., J.M.P.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Aksharkumar Dobariya
- Rare Brain Disorders Program, Department of Neurology (G.K., Q.M., G.A., V.J., A.D., L.B.G., J.M.P.), Department of Biochemistry (H.C., B.P.), Department of Pathology (J.Y.P.), Department of Physiology (J.M.P.), Department of Pediatrics (J.M.P.), and Eugene McDermott Center for Human Growth & Development/Center for Human Genetics (J.Y.P., J.M.P.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Levi B Good
- Rare Brain Disorders Program, Department of Neurology (G.K., Q.M., G.A., V.J., A.D., L.B.G., J.M.P.), Department of Biochemistry (H.C., B.P.), Department of Pathology (J.Y.P.), Department of Physiology (J.M.P.), Department of Pediatrics (J.M.P.), and Eugene McDermott Center for Human Growth & Development/Center for Human Genetics (J.Y.P., J.M.P.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Bruce Posner
- Rare Brain Disorders Program, Department of Neurology (G.K., Q.M., G.A., V.J., A.D., L.B.G., J.M.P.), Department of Biochemistry (H.C., B.P.), Department of Pathology (J.Y.P.), Department of Physiology (J.M.P.), Department of Pediatrics (J.M.P.), and Eugene McDermott Center for Human Growth & Development/Center for Human Genetics (J.Y.P., J.M.P.), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jason Y Park
- Rare Brain Disorders Program, Department of Neurology (G.K., Q.M., G.A., V.J., A.D., L.B.G., J.M.P.), Department of Biochemistry (H.C., B.P.), Department of Pathology (J.Y.P.), Department of Physiology (J.M.P.), Department of Pediatrics (J.M.P.), and Eugene McDermott Center for Human Growth & Development/Center for Human Genetics (J.Y.P., J.M.P.), University of Texas Southwestern Medical Center, Dallas, Texas.
| | - Juan M Pascual
- Rare Brain Disorders Program, Department of Neurology (G.K., Q.M., G.A., V.J., A.D., L.B.G., J.M.P.), Department of Biochemistry (H.C., B.P.), Department of Pathology (J.Y.P.), Department of Physiology (J.M.P.), Department of Pediatrics (J.M.P.), and Eugene McDermott Center for Human Growth & Development/Center for Human Genetics (J.Y.P., J.M.P.), University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
6
|
Metabolism of Exogenous [2,4- 13C]β-Hydroxybutyrate following Traumatic Brain Injury in 21-22-Day-Old Rats: An Ex Vivo NMR Study. Metabolites 2022; 12:metabo12080710. [PMID: 36005582 PMCID: PMC9414923 DOI: 10.3390/metabo12080710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/15/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023] Open
Abstract
Traumatic brain injury (TBI) is leading cause of morbidity in young children. Acute dysregulation of oxidative glucose metabolism within the first hours after injury is a hallmark of TBI. The developing brain relies on ketones as well as glucose for energy. Thus, the aim of this study was to determine the metabolism of ketones early after TBI injury in the developing brain. Following the controlled cortical impact injury model of TBI, 21-22-day-old rats were infused with [2,4-13C]β-hydroxybutyrate during the acute (4 h) period after injury. Using ex vivo 13C-NMR spectroscopy, we determined that 13C-β-hydroxybutyrate (13C-BHB) metabolism was increased in both the ipsilateral and contralateral sides of the brain after TBI. Incorporation of the label was significantly higher in glutamate than glutamine, indicating that 13C-BHB metabolism was higher in neurons than astrocytes in both sham and injured brains. Our results show that (i) ketone metabolism was significantly higher in both the ipsilateral and contralateral sides of the injured brain after TBI; (ii) ketones were extensively metabolized by both astrocytes and neurons, albeit higher in neurons; (iii) the pyruvate recycling pathway determined by incorporation of the label from the metabolism of 13C-BHB into lactate was upregulated in the immature brain after TBI.
Collapse
|
7
|
Cai M, Wang H, Song H, Yang R, Wang L, Xue X, Sun W, Hu J. Lactate Is Answerable for Brain Function and Treating Brain Diseases: Energy Substrates and Signal Molecule. Front Nutr 2022; 9:800901. [PMID: 35571940 PMCID: PMC9099001 DOI: 10.3389/fnut.2022.800901] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/24/2021] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
Research to date has provided novel insights into lactate's positive role in multiple brain functions and several brain diseases. Although notable controversies and discrepancies remain, the neurobiological role and the metabolic mechanisms of brain lactate have now been described. A theoretical framework on the relevance between lactate and brain function and brain diseases is presented. This review begins with the source and route of lactate formation in the brain and food; goes on to uncover the regulatory effect of lactate on brain function; and progresses to gathering the application and concentration variation of lactate in several brain diseases (diabetic encephalopathy, Alzheimer's disease, stroke, traumatic brain injury, and epilepsy) treatment. Finally, the dual role of lactate in the brain is discussed. This review highlights the biological effect of lactate, especially L-lactate, in brain function and disease studies and amplifies our understanding of past research.
Collapse
Affiliation(s)
- Ming Cai
- Department of Rehabilitation Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
| | - Hongbiao Wang
- Department of Physical Education, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Haihan Song
- Central Lab, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Ruoyu Yang
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Liyan Wang
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Xiangli Xue
- Key Laboratory of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Wanju Sun
- Central Lab, Shanghai Pudong New Area People's Hospital, Shanghai, China
- *Correspondence: Wanju Sun
| | - Jingyun Hu
- Central Lab, Shanghai Pudong New Area People's Hospital, Shanghai, China
- Jingyun Hu
| |
Collapse
|
8
|
Mitroshina EV, Savyuk MO, Ponimaskin E, Vedunova MV. Hypoxia-Inducible Factor (HIF) in Ischemic Stroke and Neurodegenerative Disease. Front Cell Dev Biol 2021; 9:703084. [PMID: 34395432 PMCID: PMC8355741 DOI: 10.3389/fcell.2021.703084] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/30/2021] [Accepted: 07/05/2021] [Indexed: 01/09/2023] Open
Abstract
Hypoxia is one of the most common pathological conditions, which can be induced by multiple events, including ischemic injury, trauma, inflammation, tumors, etc. The body's adaptation to hypoxia is a highly important phenomenon in both health and disease. Most cellular responses to hypoxia are associated with a family of transcription factors called hypoxia-inducible factors (HIFs), which induce the expression of a wide range of genes that help cells adapt to a hypoxic environment. Basic mechanisms of adaptation to hypoxia, and particularly HIF functions, have being extensively studied over recent decades, leading to the 2019 Nobel Prize in Physiology or Medicine. Based on their pivotal physiological importance, HIFs are attracting increasing attention as a new potential target for treating a large number of hypoxia-associated diseases. Most of the experimental work related to HIFs has focused on roles in the liver and kidney. However, increasing evidence clearly demonstrates that HIF-based responses represent an universal adaptation mechanism in all tissue types, including the central nervous system (CNS). In the CNS, HIFs are critically involved in the regulation of neurogenesis, nerve cell differentiation, and neuronal apoptosis. In this mini-review, we provide an overview of the complex role of HIF-1 in the adaptation of neurons and glia cells to hypoxia, with a focus on its potential involvement into various neuronal pathologies and on its possible role as a novel therapeutic target.
Collapse
Affiliation(s)
- Elena V. Mitroshina
- Department of Neurotechnologe, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, Nizhny Novgorod, Russia
| | - Maria O. Savyuk
- Department of Neurotechnologe, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, Nizhny Novgorod, Russia
| | - Evgeni Ponimaskin
- Department of Neurotechnologe, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, Nizhny Novgorod, Russia
- Department of Cellular Neurophysiology, Hannover Medical School, Hanover, Germany
| | - Maria V. Vedunova
- Department of Neurotechnologe, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, Nizhny Novgorod, Russia
| |
Collapse
|
9
|
Yuan J, Botchway BOA, Zhang Y, Wang X, Liu X. Combined bioscaffold with stem cells and exosomes can improve traumatic brain injury. Stem Cell Rev Rep 2021; 16:323-334. [PMID: 31808037 DOI: 10.1007/s12015-019-09927-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/21/2022]
Abstract
The intricacy of the brain, along with the existence of blood brain barrier (BBB) does complicate the delivery of effective therapeutics through simple intravascular injection. Hence, an effective delivery mechanism of therapeutics in the event of either traumatic brain injury (TBI) or other brain injuries is needed. Stem cells can promote regeneration and repair injury. The usage of biomaterials and exosomes in transporting stem cells to target lesion sites has been suggested as a potential option. The combination of biomaterials with modified exosomes can help in transporting stem cells to injury sites, whiles also increasing their survival and promoting effective treatment. Herein, we review the current researches pertinent to biological scaffolds and exosomes in repairing TBI and present the current progress and new direction in the clinical setting. We begin with the role of bioscaffold in treating neuronal conditions, the effect of exosomes in injury, and conclude with the improvement of TBI via the employment of combined exosomes, bioscaffold and stem cells.
Collapse
Affiliation(s)
- Jiaying Yuan
- Department of Histology and Embryology, Medical College, Shaoxing University, 312000, Shaoxing, Zhejiang, China
| | - Benson O A Botchway
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Yong Zhang
- Department of Histology and Embryology, Medical College, Shaoxing University, 312000, Shaoxing, Zhejiang, China
| | - Xizhi Wang
- Department of Histology and Embryology, Medical College, Shaoxing University, 312000, Shaoxing, Zhejiang, China
| | - Xuehong Liu
- Department of Histology and Embryology, Medical College, Shaoxing University, 312000, Shaoxing, Zhejiang, China.
| |
Collapse
|
10
|
Wang H, Chen J, Gao C, Chen W, Chen G, Zhang M, Luo C, Wang T, Chen X, Tao L. TMT-based proteomics analysis to screen potential biomarkers of acute-phase TBI in rats. Life Sci 2020; 264:118631. [PMID: 33131748 DOI: 10.1016/j.lfs.2020.118631] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/25/2020] [Revised: 10/09/2020] [Accepted: 10/17/2020] [Indexed: 01/19/2023]
Abstract
AIMS Traumatic brain injury (TBI) is a common nervous system injury. However, the detailed mechanisms about functional dysregulation and dignostic biomarkers post-TBI are still unclear. So we aimed to identify potential differentially expressed proteins and genes in TBI for clinical diagnosis and therapeutic purposes. MAIN METHODS Rat TBI model was established by the weight-drop method. First, through TMT-proteomics, we screened for the change in the proteins expression profile acute phase post-TBI. The DAVID and Reactome databases were used to analyze and visualize the dysregulation proteins. Then, using publicly available microarray datasets GSE45997, differentially expressed genes (DGEs) were identified for the 24 h post-TBI stage. Also, the proteomic data were compared with microarray data to analyze the similarity. KEY FINDINGS We found significant proteomics and transcriptomic changes in post-TBI samples. 989, 881, 832, 1057 proteins were quantitated at 1 h, 6 h, 24 h, and 3 d post-injury correspondingly. Concerning proteomics findings, oxygen transport, acute-phase response, and negative regulation of endopeptidase activity were influenced throughout the acute phrase of TBI. Also, pathways related to scavenging of heme from plasma, binding, and uptake of ligands by scavenger receptors were highly enriched in all time-points of TBI samples. SIGNIFICANCE We noticed that the interaction-networks trend to get complicated with more node connections following the progression of TBI. We inferred that Hk-1, PRKAR2A, and MBP could be novel candidate biomarkers related to time-injury in acute-phase TBI. Also, Ceruloplasmin and Complement C3 were found to be important proteins and genes are involved in the TBI.
Collapse
Affiliation(s)
- Haochen Wang
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou 215123, China.
| | - Jie Chen
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou 215123, China
| | - Cheng Gao
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou 215123, China
| | - Wei Chen
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou 215123, China
| | - Guang Chen
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou 215123, China
| | - Mingyang Zhang
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou 215123, China
| | - Chengliang Luo
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou 215123, China
| | - Tao Wang
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou 215123, China
| | - Xiping Chen
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou 215123, China.
| | - Luyang Tao
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou 215123, China.
| |
Collapse
|
11
|
Precision Medicine in Acute Brain Injury: A Narrative Review. J Neurosurg Anesthesiol 2020; 34:e14-e23. [PMID: 32590476 DOI: 10.1097/ana.0000000000000710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/11/2020] [Accepted: 05/24/2020] [Indexed: 11/26/2022]
Abstract
Over the past few years, the concept of personalized medicine has percolated into the management of different neurological conditions. Improving outcomes after acute brain injury (ABI) continues to be a major challenge. Unrecognized individual multiomic variations in addition to multiple interacting processes may explain why we fail to observe comprehensive improvements in ABI outcomes even when applied treatments appear to be beneficial logically. The provision of clinical care based on a multiomic approach may revolutionize the management of traumatic brain injury, delayed cerebral ischemia after subarachnoid hemorrhage, acute ischemic stroke, and several other neurological diseases. The challenge is to incorporate all the information obtained from genomic studies, other omic data, and individual variability into a practical tool that can be used to assist clinical decision-making. The effective execution of such strategies, which is still far away, requires the development of protocols on the basis of these complex interactions and strict adherence to management protocols. In this review, we will discuss various omics and physiological targets to guide individualized patient management after ABI.
Collapse
|
12
|
Zhai X, Li J, Li L, Sun Y, Zhang X, Xue Y, Lv J, Gao Y, Li S, Yan W, Yin S, Xiao Z. L-lactate preconditioning promotes plasticity-related proteins expression and reduces neurological deficits by potentiating GPR81 signaling in rat traumatic brain injury model. Brain Res 2020; 1746:146945. [PMID: 32531223 DOI: 10.1016/j.brainres.2020.146945] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/28/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 01/16/2023]
Abstract
Currently, there is no efficacious pharmacological treatment for traumatic brain injury (TBI). Previous studies revealed that L-lactate preconditioning has shown rich neuroprotective effects against cerebral ischemia, and therefore has the potential to improve neurological outcomes after TBI. L-lactate played a neuroprotective role by activating GPR81 in diseases of the central nervous system (CNS) such as TBI and cerebral ischemia. In this study we investigated the effects of L-lactate preconditioning on TBI and explored the underlying mechanisms. In this study, the mNSS test revealed that L-lactate preconditioning alleviates the neurological deficit caused by TBI in rats. L-lactate preconditioning significantly increased the expression of GPR81, PSD95, GAP43, BDNF, and MCT2 24 h after TBI in the cortex and hippocampus compared with the sham group. Taken together, these data suggested that L-lactate preconditioning is an effective method with which to recover neurological function after TBI. This reveals the mechanism of L-lactate preconditioning on TBI and provides a potential therapeutic method for TBI in humans.
Collapse
Affiliation(s)
- Xiuli Zhai
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China
| | - Jinying Li
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China
| | - Liya Li
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China
| | - Ye Sun
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China
| | - Xiaonan Zhang
- Department of Physiology, Dalian Medical University, Dalian 116044, China
| | - Ying Xue
- Department of Physiology, Dalian Medical University, Dalian 116044, China
| | - Jiaxin Lv
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Ye Gao
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China
| | - Shouxin Li
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China
| | - Wei Yan
- Department of Physiology, Dalian Medical University, Dalian 116044, China
| | - Shengming Yin
- Department of Physiology, Dalian Medical University, Dalian 116044, China.
| | - Zhaoyang Xiao
- Department of Anesthesiology, The Second Affiliated Hospital of Dalian Medical University, Dalian 116027, China.
| |
Collapse
|
13
|
Wu X, Wang C, Wang J, Zhu M, Yao Y, Liu J. Hypoxia preconditioning protects neuronal cells against traumatic brain injury through stimulation of glucose transport mediated by HIF-1α/GLUTs signaling pathway in rat. Neurosurg Rev 2020; 44:411-422. [PMID: 31897883 PMCID: PMC7851104 DOI: 10.1007/s10143-019-01228-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/05/2019] [Revised: 12/04/2019] [Accepted: 12/18/2019] [Indexed: 11/25/2022]
Abstract
Hypoxia preconditioning (HPC), a well-established preconditioning model, has been shown to protect the brain against severe hypoxia or ischemia caused by traumatic brain injury (TBI), but the mechanism has not been well elucidated. Anaerobic glycolysis is the major way for neurons to produce energy under cerebral ischemia and hypoxia after TBI, and it requires large amounts of glucose. We hypothesized that glucose transport, as a rate-limiting step of glucose metabolism, may play key roles in the neuroprotective effects of HPC on cerebral cortex tissue against TBI. The aim of this study was to investigate the effect of HPC on glucose transport activity of rat cerebral cortex tissue after TBI through examining the gene expression of two major glucose transporters (GLUT1 and GLUT3) and their upstream target gene hypoxia-inducible factor-1α (HIF-1α). Sprague-Dawley rats were treated with HPC (50.47 kPa, 3 h/d, 3d). Twenty-four hours after the last treatment, the rats were injured using the Feeney free falling model. Cortex tissues of injured rats were removed at 1 h, 4 h, 8 h, 12 h, 1 day, 3 days, 7 d, and 14 days post-injury for histological analysis. Compared with TBI alone, HPC before TBI resulted in the expression of HIF-1α, GLUT1, and GLUT3 to increase at 1 h; they were markedly increased at 4 h, 8 h, 12 h, 1 day, and 3 days and decreased thereafter (p < 0.05). HPC before TBI could improve neuronal survival in rats by examining NeuN staining and observing reduced apoptosis by examining TUNEL staining. The result showed that HPC before TBI could increase the expression of GLUT1 and GLUT3. And through double immunofluorescence staining for GLUT3 and NeuN, the results strongly suggest that HPC improved glucose transport activity of neurons in rats with TBI. In summary, our results further support that HPC can improve hypoxia tolerance and attenuate neuronal loss of cerebral cortex in rats after TBI. The mechanism is mainly related to the increase of glucose transport activity through inducing GLUT1 and GLUT3 expression through upregulating HIF-1α expression.
Collapse
Affiliation(s)
- Xiaogang Wu
- Department of Neurosurgery, No. 901 Hospital of the Chinese People's Liberation Army Logistic Support Force, Hefei, Anhui, China
| | - Chunlin Wang
- Department of Neurosurgery, No. 901 Hospital of the Chinese People's Liberation Army Logistic Support Force, Hefei, Anhui, China
| | - Jinbiao Wang
- Department of Neurosurgery, No. 901 Hospital of the Chinese People's Liberation Army Logistic Support Force, Hefei, Anhui, China
| | - Meijie Zhu
- Department of Neurosurgery, No. 901 Hospital of the Chinese People's Liberation Army Logistic Support Force, Hefei, Anhui, China
| | - Yinsheng Yao
- Department of Neurosurgery, No. 901 Hospital of the Chinese People's Liberation Army Logistic Support Force, Hefei, Anhui, China
| | - Jiachuan Liu
- Department of Neurosurgery, No. 901 Hospital of the Chinese People's Liberation Army Logistic Support Force, Hefei, Anhui, China.
| |
Collapse
|
14
|
Huynh LM, Burns MP, Taub DD, Blackman MR, Zhou J. Chronic Neurobehavioral Impairments and Decreased Hippocampal Expression of Genes Important for Brain Glucose Utilization in a Mouse Model of Mild TBI. Front Endocrinol (Lausanne) 2020; 11:556380. [PMID: 33071972 PMCID: PMC7531511 DOI: 10.3389/fendo.2020.556380] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 04/28/2020] [Accepted: 08/17/2020] [Indexed: 11/17/2022] Open
Abstract
Glucose is an essential cellular fuel for maintaining normal brain functions. Traumatic brain injury (TBI) decreases brain glucose utilization in both human and experimental animals during the acute or subacute phase of TBI. It remains unclear as to how the damages affect brain glucose utilization and its association with persistent neurobehavioral impairments in the chronic phase of mild TBI (mTBI). Accordingly, we compared expression of selected genes important to brain glucose utilization in different brain regions of mice during the chronic phase in mTBI vs. sham operated mice. These genes included hexokinase-1 (HK1), phosphofructokinase (PFK), pyruvate kinase (PK), pyruvate dehydrogenase (PDH), capillary glucose transporter (Glut-1), neuron glucose transporter (Glut-3), astrocyte lactate transpor1 (MCT-1), neuron lactate transporter (MCT-2), lactate receptor (GPR81), and Hexokinase isoform-2 (HK2). Young adult male C57BL/6J mice were brain injured with repetitive closed-head concussions. Morris water maze (MWM), elevated plus maze (EPM), and neurological severity score test (NSS) were performed for evaluation of mice neurobehavioral impairments at 2, 4, and 6 months post mTBI. Two days after completion of the last behavioral test, the frontal cortex, hippocampus, brainstem, hypothalamus, and cerebellum were collected for gene expression measurements. The expression of the mRNAs encoding PK, and PDH, two critical enzymes in glucose metabolism, was decreased at all-time points only in the hippocampus, but was unchanged in the brainstem, hypothalamus, and cortex in mTBI mice. mTBI mice also exhibited the following behavioral alterations: (1) decreased spatial learning and memory 2, 4, and 6 months after the injury, (2) increased proportion of time spent on open vs. closed arms determined by EPM, and (3) accelerated reduction in motor activity observed at 4 months, two months earlier than observed in the sham group, during the EPM testing. There were no significant differences in NSS between injury and sham groups at any of the three time points. Thus, mTBI in male mice led to persistent decreased hippocampal expression of mRNAs that encode critical glucose utilization related enzymes in association with long-term impairments in selected neurobehavioral outcomes.
Collapse
Affiliation(s)
- Linda M. Huynh
- Research Service, Washington DC VA Medical Center, Washington, DC, United States
| | - Mark P. Burns
- Department of Neuroscience, Georgetown University School of Medicine, Washington, DC, United States
| | - Daniel D. Taub
- Research Service, Washington DC VA Medical Center, Washington, DC, United States
- Department of Biochemistry and Molecular and Cell Biology, Georgetown University School of Medicine, Washington, DC, United States
| | - Marc R. Blackman
- Research Service, Washington DC VA Medical Center, Washington, DC, United States
- Department of Medicine, George Washington University School of Medicine, Washington, DC, United States
- Department of Medicine, Georgetown University School of Medicine, Washington, DC, United States
| | - June Zhou
- Research Service, Washington DC VA Medical Center, Washington, DC, United States
- *Correspondence: June Zhou
| |
Collapse
|
15
|
Pyruvate Dehydrogenase and Tricarboxylic Acid Cycle Enzymes Are Sensitive Targets of Traumatic Brain Injury Induced Metabolic Derangement. Int J Mol Sci 2019; 20:ijms20225774. [PMID: 31744143 PMCID: PMC6888669 DOI: 10.3390/ijms20225774] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/13/2019] [Revised: 11/05/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023] Open
Abstract
Using a closed-head impact acceleration model of mild or severe traumatic brain injury (mTBI or sTBI, respectively) in rats, we evaluated the effects of graded head impacts on the gene and protein expressions of pyruvate dehydrogenase (PDH), as well as major enzymes of mitochondrial tricarboxylic acid cycle (TCA). TBI was induced in anaesthetized rats by dropping 450 g from 1 (mTBI) or 2 m height (sTBI). After 6 h, 12 h, 24 h, 48 h, and 120 h gene expressions of enzymes and subunits of PDH. PDH kinases and phosphatases (PDK1-4 and PDP1-2, respectively), citrate synthase (CS), isocitrate dehydrogenase (IDH), oxoglutarate dehydrogenase (OGDH), succinate dehydrogenase (SDH), succinyl-CoA synthase (SUCLG), and malate dehydrogenase (MDH) were determined in whole brain extracts (n = 6 rats at each time for both TBI levels). In the same samples, the high performance liquid chromatographic (HPLC) determination of acetyl-coenzyme A (acetyl-CoA) and free coenzyme A (CoA-SH) was performed. Sham-operated animals (n = 6) were used as controls. After mTBI, the results indicated a general transient decrease, followed by significant increases, in PDH and TCA gene expressions. Conversely, permanent PDH and TCA downregulation occurred following sTBI. The inhibitory conditions of PDH (caused by PDP1-2 downregulations and PDK1-4 overexpression) and SDH appeared to operate only after sTBI. This produced almost no change in acetyl-CoA and free CoA-SH following mTBI and a remarkable depletion of both compounds after sTBI. These results again demonstrated temporary or steady mitochondrial malfunctioning, causing minimal or profound modifications to energy-related metabolites, following mTBI or sTBI, respectively. Additionally, PDH and SDH appeared to be highly sensitive to traumatic insults and are deeply involved in mitochondrial-related energy metabolism imbalance.
Collapse
|
16
|
Staib-Lasarzik I, Nagel N, Sebastiani A, Griemert EV, Thal SC. Analgesic treatment limits surrogate parameters for early stress and pain response after experimental subarachnoid hemorrhage. BMC Neurosci 2019; 20:49. [PMID: 31533626 PMCID: PMC6751841 DOI: 10.1186/s12868-019-0531-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/21/2019] [Accepted: 09/05/2019] [Indexed: 12/30/2022] Open
Abstract
Background In animal research, authorities require a classification of anticipated pain levels and a perioperative analgesia protocol prior to approval of the experiments. However, data on this topic is rare and so is the reported use of analgesics. We determined surrogate parameters of pain and general well-being after subarachnoid hemorrhage (SAH), as well as the potential for improvement by different systemic analgesia paradigms. Brain injury was induced by filament perforation to mimic SAH. Sham-operated mice were included as surgical control groups with either neck or no-neck preparation. Mice with controlled cortical impact (CCI) injury were included as a control group with traumatic brain injury (TBI), but without neck preparation. Mice were randomized to buprenorphine, carprofen, meloxicam, or vehicle treatment. 24 h after SAH, CCI or sham surgery, pain and stress levels were assessed with a visual assessment score and the amount of food intake was recorded. Results Neck preparation, which is required to expose the surgical field for SAH induction, already increased pain/stress levels and sham surgeries for both CCI and SAH reduced food intake. Pain/stress levels were higher and food intake was lower after SAH compared with CCI. Pain/stress levels after CCI without analgesic treatment were similar to levels after SAH sham surgery. Pain treatment with buprenorphine was effective to reduce pain after SAH, whereas lower pain/stress intensity levels after CCI were not improved. Conclusion This study emphasizes the importance of pain and stress assessment after surgeries and the efficacy of buprenorphine to improve pain and comfort levels after experimental SAH.
Collapse
Affiliation(s)
- Irina Staib-Lasarzik
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Nadine Nagel
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Anne Sebastiani
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Eva-Verena Griemert
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Serge C Thal
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, 55131, Mainz, Germany.
| |
Collapse
|
17
|
Pioglitazone improves working memory performance when administered in chronic TBI. Neurobiol Dis 2019; 132:104611. [PMID: 31513844 DOI: 10.1016/j.nbd.2019.104611] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/28/2019] [Revised: 09/02/2019] [Accepted: 09/06/2019] [Indexed: 01/26/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of long-term disability in the United States. Even in comparatively mild injuries, cognitive and behavioral symptoms can persist for years, and there are currently no established strategies for mitigating symptoms in chronic injury. A key feature of TBI-induced damage in acute and chronic injury is disruption of metabolic pathways. As neurotransmission, and therefore cognition, are highly dependent on the supply of energy, we hypothesized that modulating metabolic activity could help restore behavioral performance even when treatment was initiated weeks after TBI. We treated rats with pioglitazone, a FDA-approved drug for diabetes, beginning 46 days after lateral fluid percussion injury and tested working memory performance in the radial arm maze (RAM) after 14 days of treatment. Pioglitazone treated TBI rats performed significantly better in the RAM test than untreated TBI rats, and similarly to control animals. While hexokinase activity in hippocampus was increased by pioglitazone treatment, there was no upregulation of either the neuronal glucose transporter or hexokinase enzyme expression. Expression of glial markers GFAP and Iba-1 were also not influenced by pioglitazone treatment. These studies suggest that targeting brain metabolism, in particular hippocampal metabolism, may be effective in alleviating cognitive symptoms in chronic TBI.
Collapse
|
18
|
Royes LFF, Gomez-Pinilla F. Making sense of gut feelings in the traumatic brain injury pathogenesis. Neurosci Biobehav Rev 2019; 102:345-361. [PMID: 31102601 DOI: 10.1016/j.neubiorev.2019.05.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/15/2019] [Revised: 05/13/2019] [Accepted: 05/14/2019] [Indexed: 02/06/2023]
Abstract
Traumatic brain injury (TBI) is a devastating condition which often initiates a sequel of neurological disorders that can last throughout lifespan. From metabolic perspective, TBI also compromises systemic physiology including the function of body organs with subsequent malfunctions in metabolism. The emerging panorama is that the effects of TBI on the periphery strike back on the brain and exacerbate the overall TBI pathogenesis. An increasing number of clinical reports are alarming to show that metabolic dysfunction is associated with incidence of long-term neurological and psychiatric disorders. The autonomic nervous system, associated hypothalamic-pituitary axis, and the immune system are at the center of the interface between brain and body and are central to the regulation of overall homeostasis and disease. We review the strong association between mechanisms that regulate cell metabolism and inflammation which has important clinical implications for the communication between body and brain. We also discuss the integrative actions of lifestyle interventions such as diet and exercise on promoting brain and body health and cognition after TBI.
Collapse
Affiliation(s)
- Luiz Fernando Freire Royes
- Exercise Biochemistry Laboratory, Center of Physical Education and Sports, Federal University of Santa Maria - UFSM, Santa Maria, RS, Brazil
| | - Fernando Gomez-Pinilla
- Departments of Neurosurgery, and Integrative and Biology and Physiology, UCLA Brain Injury Research Center, University of California, Los Angeles, USA.
| |
Collapse
|
19
|
Koenig JB, Cantu D, Low C, Sommer M, Noubary F, Croker D, Whalen M, Kong D, Dulla CG. Glycolytic inhibitor 2-deoxyglucose prevents cortical hyperexcitability after traumatic brain injury. JCI Insight 2019; 5:126506. [PMID: 31038473 DOI: 10.1172/jci.insight.126506] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/31/2022] Open
Abstract
Traumatic brain injury (TBI) causes cortical dysfunction and can lead to post-traumatic epilepsy. Multiple studies demonstrate that GABAergic inhibitory network function is compromised following TBI, which may contribute to hyperexcitability and motor, behavioral, and cognitive deficits. Preserving the function of GABAergic interneurons, therefore, is a rational therapeutic strategy to preserve cortical function after TBI and prevent long-term clinical complications. Here, we explored an approach based on the ketogenic diet, a neuroprotective and anticonvulsant dietary therapy which results in reduced glycolysis and increased ketosis. Utilizing a pharmacologic inhibitor of glycolysis (2-deoxyglucose, or 2-DG), we found that acute in vitro application of 2-DG decreased the excitability of excitatory neurons, but not inhibitory interneurons, in cortical slices from naïve mice. Employing the controlled cortical impact (CCI) model of TBI in mice, we found that in vitro 2-DG treatment rapidly attenuated epileptiform activity seen in acute cortical slices 3 to 5 weeks after TBI. One week of in vivo 2-DG treatment immediately after TBI prevented the development of epileptiform activity, restored excitatory and inhibitory synaptic activity, and attenuated the loss of parvalbumin-expressing inhibitory interneurons. In summary, 2-DG may have therapeutic potential to restore network function following TBI.
Collapse
Affiliation(s)
- Jenny B Koenig
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA.,Neuroscience Program, Tufts University Sackler School of Graduate Biomedical Sciences, Boston, Massachusetts, USA
| | - David Cantu
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Cho Low
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA.,Cellular, Molecular, and Developmental Biology Program, Tufts University Sackler School of Graduate Biomedical Sciences, Boston, Massachusetts, USA
| | - Mary Sommer
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Farzad Noubary
- Department of Health Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Danielle Croker
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Michael Whalen
- Neuroscience Center, Harvard Medical School, Massachusetts General Hospital, Charlestown, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Dong Kong
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Koenig JB, Dulla CG. Dysregulated Glucose Metabolism as a Therapeutic Target to Reduce Post-traumatic Epilepsy. Front Cell Neurosci 2018; 12:350. [PMID: 30459556 PMCID: PMC6232824 DOI: 10.3389/fncel.2018.00350] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/19/2018] [Accepted: 09/19/2018] [Indexed: 12/13/2022] Open
Abstract
Traumatic brain injury (TBI) is a significant cause of disability worldwide and can lead to post-traumatic epilepsy. Multiple molecular, cellular, and network pathologies occur following injury which may contribute to epileptogenesis. Efforts to identify mechanisms of disease progression and biomarkers which predict clinical outcomes have focused heavily on metabolic changes. Advances in imaging approaches, combined with well-established biochemical methodologies, have revealed a complex landscape of metabolic changes that occur acutely after TBI and then evolve in the days to weeks after. Based on this rich clinical and preclinical data, combined with the success of metabolic therapies like the ketogenic diet in treating epilepsy, interest has grown in determining whether manipulating metabolic activity following TBI may have therapeutic value to prevent post-traumatic epileptogenesis. Here, we focus on changes in glucose utilization and glycolytic activity in the brain following TBI and during seizures. We review relevant literature and outline potential paths forward to utilize glycolytic inhibitors as a disease-modifying therapy for post-traumatic epilepsy.
Collapse
Affiliation(s)
- Jenny B Koenig
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|