51
|
Yonutas HM, Vekaria HJ, Sullivan PG. Mitochondrial specific therapeutic targets following brain injury. Brain Res 2016; 1640:77-93. [PMID: 26872596 DOI: 10.1016/j.brainres.2016.02.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 01/29/2016] [Accepted: 02/02/2016] [Indexed: 02/03/2023]
Abstract
Traumatic brain injury is a complicated disease to treat due to the complex multi-factorial secondary injury cascade that is initiated following the initial impact. This secondary injury cascade causes nonmechanical tissue damage, which is where therapeutic interventions may be efficacious for intervention. One therapeutic target that has shown much promise following brain injury are mitochondria. Mitochondria are complex organelles found within the cell. At a superficial level, mitochondria are known to produce the energy substrate used within the cell called ATP. However, their importance to overall cellular homeostasis is even larger than their production of ATP. These organelles are necessary for calcium cycling, ROS production and play a role in the initiation of cell death pathways. When mitochondria become dysfunctional, they can become dysregulated leading to a loss of cellular homeostasis and eventual cell death. Within this review there will be a deep discussion into mitochondrial bioenergetics followed by a brief discussion into traumatic brain injury and how mitochondria play an integral role in the neuropathological sequelae following an injury. The review will conclude with a discussion pertaining to the therapeutic approaches currently being studied to ameliorate mitochondrial dysfunction following brain injury. This article is part of a Special Issue entitled SI:Brain injury and recovery.
Collapse
Affiliation(s)
- H M Yonutas
- University of Kentucky, 741 South Limestone Street, BBSRB 475, 30536 Lexington, United States
| | - H J Vekaria
- University of Kentucky, 741 South Limestone Street, BBSRB 475, 30536 Lexington, United States
| | - P G Sullivan
- University of Kentucky, 741 South Limestone Street, BBSRB 475, 30536 Lexington, United States.
| |
Collapse
|
52
|
Stefano GB, Kream RM. Dysregulated mitochondrial and chloroplast bioenergetics from a translational medical perspective (Review). Int J Mol Med 2016; 37:547-55. [PMID: 26821064 PMCID: PMC4771107 DOI: 10.3892/ijmm.2016.2471] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 01/22/2016] [Indexed: 02/06/2023] Open
Abstract
Mitochondria and chloroplasts represent endosymbiotic models of complex organelle development, driven by intense evolutionary pressure to provide exponentially enhanced ATP-dependent energy production functionally linked to cellular respiration and photosynthesis. Within the realm of translational medicine, it has become compellingly evident that mitochondrial dysfunction, resulting in compromised cellular bioenergetics, represents a key causative factor in the etiology and persistence of major diseases afflicting human populations. As a pathophysiological consequence of enhanced oxygen utilization that is functionally uncoupled from the oxidative phosphorylation of ADP, significant levels of reactive oxygen species (ROS) may be generated within mitochondria and chloroplasts, which may effectively compromise cellular energy production following prolonged stress/inflammatory conditions. Empirically determined homologies in biochemical pathways, and their respective encoding gene sequences between chloroplasts and mitochondria, suggest common origins via entrapped primordial bacterial ancestors. From evolutionary and developmental perspectives, the elucidation of multiple biochemical and molecular relationships responsible for errorless bioenergetics within mitochondrial and plastid complexes will most certainly enhance the depth of translational approaches to ameliorate or even prevent the destructive effects of multiple disease states. The selective choice of discussion points contained within the present review is designed to provide theoretical bases and translational insights into the pathophysiology of human diseases from a perspective of dysregulated mitochondrial bioenergetics with special reference to chloroplast biology.
Collapse
|
53
|
Pandya JD, Sullivan PG, Leung LY, Tortella FC, Shear DA, Deng-Bryant Y. Advanced and High-Throughput Method for Mitochondrial Bioenergetics Evaluation in Neurotrauma. Methods Mol Biol 2016; 1462:597-610. [PMID: 27604740 DOI: 10.1007/978-1-4939-3816-2_32] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Mitochondrial dysfunction is one of the key posttraumatic neuropathological events observed in various experimental models of traumatic brain injury (TBI). The extent of mitochondrial dysfunction has been associated with the severity and time course of secondary injury following brain trauma. Critically, several mitochondrial targeting preclinical drugs used in experimental TBI models have shown improved mitochondrial bioenergetics, together with cortical tissue sparing and cognitive behavioral outcome. Mitochondria, being a central regulator of cellular metabolic pathways and energy producer of cells, are of a great interest for researchers aiming to adopt cutting-edge methodology for mitochondrial bioenergetics assessment. The traditional way of mitochondrial bioenergetics analysis utilizing a Clark-type oxygen electrode (aka. oxytherm) is time-consuming and labor-intensive. In the present chapter, we describe an advanced and high-throughput method for mitochondrial bioenergetics assessments utilizing the Seahorse Biosciences XF(e)24 Flux Analyzer. This allows for simultaneous measurement of multiple samples with higher efficiency than the oxytherm procedure. This chapter provides helpful guidelines for conducting mitochondrial isolation and studying mitochondrial bioenergetics in brain tissue homogenates following experimental TBI.
Collapse
Affiliation(s)
- Jignesh D Pandya
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, USA.
| | - Patrick G Sullivan
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
| | - Lai Yee Leung
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Frank C Tortella
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Deborah A Shear
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Ying Deng-Bryant
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| |
Collapse
|
54
|
Levine J, Kwon E, Paez P, Yan W, Czerwieniec G, Loo JA, Sofroniew MV, Wanner IB. Traumatically injured astrocytes release a proteomic signature modulated by STAT3-dependent cell survival. Glia 2015; 64:668-94. [PMID: 26683444 DOI: 10.1002/glia.22953] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 11/20/2015] [Indexed: 01/02/2023]
Abstract
Molecular markers associated with CNS injury are of diagnostic interest. Mechanical trauma generates cellular deformation associated with membrane permeability with unknown molecular consequences. We used an in vitro model of stretch-injury and proteomic analyses to determine protein changes in murine astrocytes and their surrounding fluids. Abrupt pressure-pulse stretching resulted in the rapid release of 59 astrocytic proteins with profiles reflecting cell injury and cell death, i.e., mechanoporation and cell lysis. This acute trauma-release proteome was overrepresented with metabolic proteins compared with the uninjured cellular proteome, bearing relevance for post-traumatic metabolic depression. Astrocyte-specific deletion of signal transducer and activator of transcription 3 (STAT3-CKO) resulted in reduced stretch-injury tolerance, elevated necrosis and increased protein release. Consistent with more lysed cells, more protein complexes, nuclear and transport proteins were released from STAT3-CKO versus nontransgenic astrocytes. STAT3-CKO astrocytes had reduced basal expression of GFAP, lactate dehydrogenase B (LDHB), aldolase C (ALDOC), and astrocytic phosphoprotein 15 (PEA15), and elevated levels of tropomyosin (TPM4) and α actinin 4 (ACTN4). Stretching caused STAT3-dependent cellular depletion of PEA15 and GFAP, and its filament disassembly in subpopulations of injured astrocytes. PEA15 and ALDOC signals were low in injured astrocytes acutely after mouse spinal cord crush injury and were robustly expressed in reactive astrocytes 1 day postinjury. In contrast, α crystallin (CRYAB) was present in acutely injured astrocytes, and absent from uninjured and reactive astrocytes, demonstrating novel marker differences among postinjury astrocytes. These findings reveal a proteomic signature of traumatically-injured astrocytes reflecting STAT3-dependent cellular survival with potential diagnostic value.
Collapse
Affiliation(s)
- Jaclynn Levine
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Eunice Kwon
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Pablo Paez
- Department of Pharmacology and Toxicology, Hunter James Kelly Research Institute, School of Medicine and Biomedical Sciences, SUNY, University at Buffalo, NYS Center of Excellence, Buffalo, New York
| | - Weihong Yan
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California
| | - Gregg Czerwieniec
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California
| | - Joseph A Loo
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California.,Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California.,UCLA/DOE Institute for Genomics and Proteomics, University of California, Los Angeles, California
| | - Michael V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Ina-Beate Wanner
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
55
|
Buonora JE, Mousseau M, Jacobowitz DM, Lazarus RC, Yarnell AM, Olsen CH, Pollard HB, Diaz-Arrastia R, Latour L, Mueller GP. Autoimmune Profiling Reveals Peroxiredoxin 6 as a Candidate Traumatic Brain Injury Biomarker. J Neurotrauma 2015; 32:1805-14. [PMID: 25938937 PMCID: PMC4651056 DOI: 10.1089/neu.2014.3736] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Autoimmune profiling in rats revealed the antioxidant enzyme, peroxiredoxin 6 (PRDX6), as a target for autoantibodies evoked in response to traumatic brain injury (TBI). Consistent with this proposal, immunohistochemical analysis of rat cerebral cortex demonstrated that PRDX6 is highly expressed in the perivascular space, presumably contained within astrocytic foot processes. Accordingly, an immunosorbent electrochemiluminescence assay was developed for investigating PRDX6 in human samples. PRDX6 was found to be measurable in human blood and highly expressed in human cerebral cortex and platelets. Circulating levels of PRDX6 were elevated fourfold over control values 4 to 24 h following mild-to-moderate TBI. These findings suggest that PRDX6 may serve as a biomarker for TBI and that autoimmune profiling is a viable strategy for the discovery of novel TBI biomarkers.
Collapse
Affiliation(s)
- John E Buonora
- 1 Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences , Bethesda, Maryland.,2 Program in Neuroscience, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Michael Mousseau
- 1 Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences , Bethesda, Maryland.,3 Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - David M Jacobowitz
- 2 Program in Neuroscience, Uniformed Services University of the Health Sciences , Bethesda, Maryland.,3 Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Rachel C Lazarus
- 2 Program in Neuroscience, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Angela M Yarnell
- 2 Program in Neuroscience, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Cara H Olsen
- 4 Biostatistics Consulting Center, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Harvey B Pollard
- 1 Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences , Bethesda, Maryland.,2 Program in Neuroscience, Uniformed Services University of the Health Sciences , Bethesda, Maryland.,3 Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Ramon Diaz-Arrastia
- 1 Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences , Bethesda, Maryland.,2 Program in Neuroscience, Uniformed Services University of the Health Sciences , Bethesda, Maryland.,5 Department of Neurology, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Lawrence Latour
- 6 Section on Stroke Diagnostics and Therapeutics, National Institute of Neurological Disorders and Stroke , Bethesda, Maryland
| | - Gregory P Mueller
- 1 Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences , Bethesda, Maryland.,2 Program in Neuroscience, Uniformed Services University of the Health Sciences , Bethesda, Maryland.,3 Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| |
Collapse
|
56
|
Harish G, Mahadevan A, Pruthi N, Sreenivasamurthy SK, Puttamallesh VN, Keshava Prasad TS, Shankar SK, Srinivas Bharath MM. Characterization of traumatic brain injury in human brains reveals distinct cellular and molecular changes in contusion and pericontusion. J Neurochem 2015; 134:156-72. [PMID: 25712633 DOI: 10.1111/jnc.13082] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 01/07/2015] [Accepted: 02/19/2015] [Indexed: 12/22/2022]
Abstract
Traumatic brain injury (TBI) contributes to fatalities and neurological disabilities worldwide. While primary injury causes immediate damage, secondary events contribute to long-term neurological defects. Contusions (Ct) are primary injuries correlated with poor clinical prognosis, and can expand leading to delayed neurological deterioration. Pericontusion (PC) (penumbra), the region surrounding Ct, can also expand with edema, increased intracranial pressure, ischemia, and poor clinical outcome. Analysis of Ct and PC can therefore assist in understanding the pathobiology of TBI and its management. This study on human TBI brains noted extensive neuronal, astroglial and inflammatory changes, alterations in mitochondrial, synaptic and oxidative markers, and associated proteomic profile, with distinct differences in Ct and PC. While Ct displayed petechial hemorrhages, thrombosis, inflammation, neuronal pyknosis, and astrogliosis, PC revealed edema, vacuolation of neuropil, axonal loss, and dystrophic changes. Proteomic analysis demonstrated altered immune response, synaptic, and mitochondrial dysfunction, among others, in Ct, while PC displayed altered regulation of neurogenesis and cytoskeletal architecture, among others. TBI brains displayed oxidative damage, glutathione depletion, mitochondrial dysfunction, and loss of synaptic proteins, with these changes being more profound in Ct. We suggest that analysis of markers specific to Ct and PC may be valuable in the evaluation of TBI pathobiology and therapeutics. We have characterized the primary injury in human traumatic brain injury (TBI). Contusions (Ct) - the injury core displayed hemorrhages, inflammation, and astrogliosis, while the surrounding pericontusion (PC) revealed edema, vacuolation, microglial activation, axonal loss, and dystrophy. Proteomic analysis demonstrated altered immune response, synaptic and mitochondrial dysfunction in Ct, and altered regulation of neurogenesis and cytoskeletal architecture in PC. Ct displayed more oxidative damage, mitochondrial, and synaptic dysfunction compared to PC.
Collapse
Affiliation(s)
- Gangadharappa Harish
- Department of Neurochemistry, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, Karnataka, India
| | - Anita Mahadevan
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, Karnataka, India
| | - Nupur Pruthi
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, Karnataka, India
| | | | | | | | - Susarla Krishna Shankar
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, Karnataka, India
| | | |
Collapse
|
57
|
Kochanek PM, Jackson TC, Ferguson NM, Carlson SW, Simon DW, Brockman EC, Ji J, Bayir H, Poloyac SM, Wagner AK, Kline AE, Empey PE, Clark RS, Jackson EK, Dixon CE. Emerging therapies in traumatic brain injury. Semin Neurol 2015; 35:83-100. [PMID: 25714870 PMCID: PMC4356170 DOI: 10.1055/s-0035-1544237] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Despite decades of basic and clinical research, treatments to improve outcomes after traumatic brain injury (TBI) are limited. However, based on the recent recognition of the prevalence of mild TBI, and its potential link to neurodegenerative disease, many new and exciting secondary injury mechanisms have been identified and several new therapies are being evaluated targeting both classic and novel paradigms. This includes a robust increase in both preclinical and clinical investigations. Using a mechanism-based approach the authors define the targets and emerging therapies for TBI. They address putative new therapies for TBI across both the spectrum of injury severity and the continuum of care, from the field to rehabilitation. They discussTBI therapy using 11 categories, namely, (1) excitotoxicity and neuronal death, (2) brain edema, (3) mitochondria and oxidative stress, (4) axonal injury, (5) inflammation, (6) ischemia and cerebral blood flow dysregulation, (7) cognitive enhancement, (8) augmentation of endogenous neuroprotection, (9) cellular therapies, (10) combination therapy, and (11) TBI resuscitation. The current golden age of TBI research represents a special opportunity for the development of breakthroughs in the field.
Collapse
Affiliation(s)
- Patrick M. Kochanek
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Travis C. Jackson
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Nikki Miller Ferguson
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Shaun W. Carlson
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departmentol Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Dennis W. Simon
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Erik C. Brockman
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jing Ji
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Hülya Bayir
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Environmental and Occupational Health, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Samuel M. Poloyac
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Environmental and Occupational Health, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Amy K. Wagner
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Anthony E. Kline
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Philip E. Empey
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Environmental and Occupational Health, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Robert S.B. Clark
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departments of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Edwin K. Jackson
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - C. Edward Dixon
- Safar Center for Resuscitation Research, University of Pittburgh School of Medicine, Pittsburgh, Pennsylvania
- Departmentol Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
58
|
Lazarus RC, Buonora JE, Jacobowitz DM, Mueller GP. Protein carbonylation after traumatic brain injury: cell specificity, regional susceptibility, and gender differences. Free Radic Biol Med 2015; 78:89-100. [PMID: 25462645 DOI: 10.1016/j.freeradbiomed.2014.10.507] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 10/07/2014] [Accepted: 10/10/2014] [Indexed: 12/15/2022]
Abstract
Protein carbonylation is a well-documented and quantifiable consequence of oxidative stress in several neuropathologies, including multiple sclerosis, Alzheimer׳s disease, and Parkinson׳s disease. Although oxidative stress is a hallmark of traumatic brain injury (TBI), little work has explored the specific neural regions and cell types in which protein carbonylation occurs. Furthermore, the effect of gender on protein carbonylation after TBI has not been studied. The present investigation was designed to determine the regional and cell specificity of TBI-induced protein carbonylation and how this response to injury is affected by gender. Immunohistochemistry was used to visualize protein carbonylation in the brains of adult male and female Sprague-Dawley rats subjected to controlled cortical impact (CCI) as an injury model of TBI. Cell-specific markers were used to colocalize the presence of carbonylated proteins in specific cell types, including astrocytes, neurons, microglia, and oligodendrocytes. Results also indicated that the injury lesion site, ventral portion of the dorsal third ventricle, and ventricular lining above the median eminence showed dramatic increases in protein carbonylation after injury. Specifically, astrocytes and limited regions of ependymal cells adjacent to the dorsal third ventricle and the median eminence were most susceptible to postinjury protein carbonylation. However, these patterns of differential susceptibility to protein carbonylation were gender dependent, with males showing significantly greater protein carbonylation at sites distant from the lesion. Proteomic analyses were also conducted and determined that the proteins most affected by carbonylation in response to TBI include glial fibrillary acidic protein, dihydropyrimidase-related protein 2, fructose-bisphosphate aldolase C, and fructose-bisphosphate aldolase A. Many other proteins, however, were not carbonylated by CCI. These findings indicate that there is both regional and protein specificity in protein carbonylation after TBI. The marked increase in carbonylation seen in ependymal layers distant from the lesion suggests a mechanism involving the transmission of a cerebral spinal fluid-borne factor to these sites. Furthermore, this process is affected by gender, suggesting that hormonal mechanisms may serve a protective role against oxidative stress.
Collapse
Affiliation(s)
- Rachel C Lazarus
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - John E Buonora
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - David M Jacobowitz
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Gregory P Mueller
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| |
Collapse
|
59
|
Dollé JP, Morrison B, Schloss RS, Yarmush ML. Brain-on-a-chip microsystem for investigating traumatic brain injury: Axon diameter and mitochondrial membrane changes play a significant role in axonal response to strain injuries. TECHNOLOGY 2014; 2:106. [PMID: 25101309 PMCID: PMC4120884 DOI: 10.1142/s2339547814500095] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Diffuse axonal injury (DAI) is a devastating consequence of traumatic brain injury, resulting in significant axon and neuronal degeneration. Currently, therapeutic options are limited. Using our brain-on-a-chip device, we evaluated axonal responses to DAI. We observed that axonal diameter plays a significant role in response to strain injury, which correlated to delayed elasticity and inversely correlated to axonal beading and axonal degeneration. When changes in mitochondrial membrane potential (MMP) were monitored an applied strain injury threshold was noted, below which delayed hyperpolarization was observed and above which immediate depolarization occurred. When the NHE-1 inhibitor EIPA was administered before injury, inhibition in both hyperpolarization and depolarization occurred along with axonal degeneration. Therefore, axonal diameter plays a significant role in strain injury and our brain-on-a-chip technology can be used both to understand the biochemical consequences of DAI and screen for potential therapeutic agents.
Collapse
|
60
|
Lama S, Auer RN, Tyson R, Gallagher CN, Tomanek B, Sutherland GR. Lactate storm marks cerebral metabolism following brain trauma. J Biol Chem 2014; 289:20200-8. [PMID: 24849602 DOI: 10.1074/jbc.m114.570978] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Brain metabolism is thought to be maintained by neuronal-glial metabolic coupling. Glia take up glutamate from the synaptic cleft for conversion into glutamine, triggering glial glycolysis and lactate production. This lactate is shuttled into neurons and further metabolized. The origin and role of lactate in severe traumatic brain injury (TBI) remains controversial. Using a modified weight drop model of severe TBI and magnetic resonance (MR) spectroscopy with infusion of (13)C-labeled glucose, lactate, and acetate, the present study investigated the possibility that neuronal-glial metabolism is uncoupled following severe TBI. Histopathology of the model showed severe brain injury with subarachnoid and hemorrhage together with glial cell activation and positive staining for Tau at 90 min post-trauma. High resolution MR spectroscopy of brain metabolites revealed significant labeling of lactate at C-3 and C-2 irrespective of the infused substrates. Increased (13)C-labeled lactate in all study groups in the absence of ischemia implied activated astrocytic glycolysis and production of lactate with failure of neuronal uptake (i.e. a loss of glial sensing for glutamate). The early increase in extracellular lactate in severe TBI with the injured neurons rendered unable to pick it up probably contributes to a rapid progression toward irreversible injury and pan-necrosis. Hence, a method to detect and scavenge the excess extracellular lactate on site or early following severe TBI may be a potential primary therapeutic measure.
Collapse
Affiliation(s)
- Sanju Lama
- From the Department of Clinical Neurosciences and the Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 2T9, Canada and
| | - Roland N Auer
- the Hôpital Ste-Justine, Département de Pathologie, Université de Montréal, Montreal, Québec H3T 1C5, Canada
| | - Randy Tyson
- From the Department of Clinical Neurosciences and the Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 2T9, Canada and
| | - Clare N Gallagher
- From the Department of Clinical Neurosciences and the Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 2T9, Canada and
| | - Boguslaw Tomanek
- From the Department of Clinical Neurosciences and the Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 2T9, Canada and
| | - Garnette R Sutherland
- From the Department of Clinical Neurosciences and the Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, Alberta T2N 2T9, Canada and
| |
Collapse
|
61
|
Patel SP, Sullivan PG, Pandya JD, Goldstein GA, VanRooyen JL, Yonutas HM, Eldahan KC, Morehouse J, Magnuson DSK, Rabchevsky AG. N-acetylcysteine amide preserves mitochondrial bioenergetics and improves functional recovery following spinal trauma. Exp Neurol 2014; 257:95-105. [PMID: 24805071 DOI: 10.1016/j.expneurol.2014.04.026] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 04/25/2014] [Accepted: 04/29/2014] [Indexed: 12/15/2022]
Abstract
Mitochondrial dysfunction is becoming a pivotal target for neuroprotective strategies following contusion spinal cord injury (SCI) and the pharmacological compounds that maintain mitochondrial function confer neuroprotection and improve long-term hindlimb function after injury. In the current study we evaluated the efficacy of cell-permeating thiol, N-acetylcysteine amide (NACA), a precursor of endogenous antioxidant glutathione (GSH), on mitochondrial function acutely, and long-term tissue sparing and hindlimb locomotor recovery following upper lumbar contusion SCI. Some designated injured adult female Sprague-Dawley rats (n=120) received either vehicle or NACA (75, 150, 300 or 600mg/kg) at 15min and 6h post-injury. After 24h the total, synaptic, and non-synaptic mitochondrial populations were isolated from a single 1.5cm spinal cord segment (centered at injury site) and assessed for mitochondrial bioenergetics. Results showed compromised total mitochondrial bioenergetics following acute SCI that was significantly improved with NACA treatment in a dose-dependent manner, with maximum effects at 300mg/kg (n=4/group). For synaptic and non-synaptic mitochondria, only 300mg/kg NACA dosage showed efficacy. Similar dosage (300mg/kg) also maintained mitochondrial GSH near normal levels. Other designated injured rats (n=21) received continuous NACA (150 or 300mg/kg/day) treatment starting at 15min post-injury for one week to assess long-term functional recovery over 6weeks post-injury. Locomotor testing and novel gait analyses showed significantly improved hindlimb function with NACA that were associated with increased tissue sparing at the injury site. Overall, NACA treatment significantly maintained acute mitochondrial bioenergetics and normalized GSH levels following SCI, and prolonged delivery resulted in significant tissue sparing and improved recovery of hindlimb function.
Collapse
Affiliation(s)
- Samir P Patel
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, Lexington, KY 40536-0509, USA
| | - Patrick G Sullivan
- Spinal Cord and Brain Injury Research Center, Department of Anatomy & Neurobiology, University of Kentucky, Lexington, KY 40536-0509, USA
| | - Jignesh D Pandya
- Spinal Cord and Brain Injury Research Center, Department of Anatomy & Neurobiology, University of Kentucky, Lexington, KY 40536-0509, USA
| | - Glenn A Goldstein
- Pediatric Endocrinology, Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Jenna L VanRooyen
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, Lexington, KY 40536-0509, USA
| | - Heather M Yonutas
- Spinal Cord and Brain Injury Research Center, Department of Anatomy & Neurobiology, University of Kentucky, Lexington, KY 40536-0509, USA
| | - Khalid C Eldahan
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, Lexington, KY 40536-0509, USA
| | - Johnny Morehouse
- Departments of Neurological Surgery, Anatomical Science, and Neurobiology, University of Louisville, Louisville, KY 40292, USA
| | - David S K Magnuson
- Departments of Neurological Surgery, Anatomical Science, and Neurobiology, University of Louisville, Louisville, KY 40292, USA
| | - Alexander G Rabchevsky
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, Lexington, KY 40536-0509, USA.
| |
Collapse
|
62
|
Patel SP, Sullivan PG, Pandya JD, Goldstein GA, VanRooyen JL, Yonutas HM, Eldahan KC, Morehouse J, Magnuson DSK, Rabchevsky AG. N-acetylcysteine amide preserves mitochondrial bioenergetics and improves functional recovery following spinal trauma. Exp Neurol 2014. [PMID: 24805071 DOI: 10.1016/j.expn eurol.2014.04.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Mitochondrial dysfunction is becoming a pivotal target for neuroprotective strategies following contusion spinal cord injury (SCI) and the pharmacological compounds that maintain mitochondrial function confer neuroprotection and improve long-term hindlimb function after injury. In the current study we evaluated the efficacy of cell-permeating thiol, N-acetylcysteine amide (NACA), a precursor of endogenous antioxidant glutathione (GSH), on mitochondrial function acutely, and long-term tissue sparing and hindlimb locomotor recovery following upper lumbar contusion SCI. Some designated injured adult female Sprague-Dawley rats (n=120) received either vehicle or NACA (75, 150, 300 or 600mg/kg) at 15min and 6h post-injury. After 24h the total, synaptic, and non-synaptic mitochondrial populations were isolated from a single 1.5cm spinal cord segment (centered at injury site) and assessed for mitochondrial bioenergetics. Results showed compromised total mitochondrial bioenergetics following acute SCI that was significantly improved with NACA treatment in a dose-dependent manner, with maximum effects at 300mg/kg (n=4/group). For synaptic and non-synaptic mitochondria, only 300mg/kg NACA dosage showed efficacy. Similar dosage (300mg/kg) also maintained mitochondrial GSH near normal levels. Other designated injured rats (n=21) received continuous NACA (150 or 300mg/kg/day) treatment starting at 15min post-injury for one week to assess long-term functional recovery over 6weeks post-injury. Locomotor testing and novel gait analyses showed significantly improved hindlimb function with NACA that were associated with increased tissue sparing at the injury site. Overall, NACA treatment significantly maintained acute mitochondrial bioenergetics and normalized GSH levels following SCI, and prolonged delivery resulted in significant tissue sparing and improved recovery of hindlimb function.
Collapse
Affiliation(s)
- Samir P Patel
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, Lexington, KY 40536-0509, USA
| | - Patrick G Sullivan
- Spinal Cord and Brain Injury Research Center, Department of Anatomy & Neurobiology, University of Kentucky, Lexington, KY 40536-0509, USA
| | - Jignesh D Pandya
- Spinal Cord and Brain Injury Research Center, Department of Anatomy & Neurobiology, University of Kentucky, Lexington, KY 40536-0509, USA
| | - Glenn A Goldstein
- Pediatric Endocrinology, Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Jenna L VanRooyen
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, Lexington, KY 40536-0509, USA
| | - Heather M Yonutas
- Spinal Cord and Brain Injury Research Center, Department of Anatomy & Neurobiology, University of Kentucky, Lexington, KY 40536-0509, USA
| | - Khalid C Eldahan
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, Lexington, KY 40536-0509, USA
| | - Johnny Morehouse
- Departments of Neurological Surgery, Anatomical Science, and Neurobiology, University of Louisville, Louisville, KY 40292, USA
| | - David S K Magnuson
- Departments of Neurological Surgery, Anatomical Science, and Neurobiology, University of Louisville, Louisville, KY 40292, USA
| | - Alexander G Rabchevsky
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, Lexington, KY 40536-0509, USA.
| |
Collapse
|
63
|
Pandya JD, Readnower RD, Patel SP, Yonutas HM, Pauly JR, Goldstein GA, Rabchevsky AG, Sullivan PG. N-acetylcysteine amide confers neuroprotection, improves bioenergetics and behavioral outcome following TBI. Exp Neurol 2014; 257:106-13. [PMID: 24792639 DOI: 10.1016/j.expneurol.2014.04.020] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 04/16/2014] [Accepted: 04/24/2014] [Indexed: 02/07/2023]
Abstract
Traumatic brain injury (TBI) has become a growing epidemic but no approved pharmacological treatment has been identified. Our previous work indicates that mitochondrial oxidative stress/damage and loss of bioenergetics play a pivotal role in neuronal cell death and behavioral outcome following experimental TBI. One tactic that has had some experimental success is to target glutathione using its precursor N-acetylcysteine (NAC). However, this approach has been hindered by the low CNS bioavailability of NAC. The current study evaluated a novel, cell permeant amide form of N-acetylcysteine (NACA), which has high permeability through cellular and mitochondrial membranes resulting in increased CNS bioavailability. Cortical tissue sparing, cognitive function and oxidative stress markers were assessed in rats treated with NACA, NAC, or vehicle following a TBI. At 15days post-injury, animals treated with NACA demonstrated significant improvements in cognitive function and cortical tissue sparing compared to NAC or vehicle treated animals. NACA treatment also was shown to reduce oxidative damage (HNE levels) at 7days post-injury. Mechanistically, post-injury NACA administration was demonstrated to maintain levels of mitochondrial glutathione and mitochondrial bioenergetics comparable to sham animals. Collectively these data provide a basic platform to consider NACA as a novel therapeutic agent for treatment of TBI.
Collapse
Affiliation(s)
- Jignesh D Pandya
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536; Department of Anatomy and Neurobiology, University of Kentucky, Lexington, KY 40536
| | - Ryan D Readnower
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536; Department of Anatomy and Neurobiology, University of Kentucky, Lexington, KY 40536
| | - Samir P Patel
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536; Department of Physiology, University of Kentucky, Lexington, KY 40536
| | - Heather M Yonutas
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536; Department of Anatomy and Neurobiology, University of Kentucky, Lexington, KY 40536
| | - James R Pauly
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536; Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536
| | - Glenn A Goldstein
- Pediatric Endocrinology Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Alexander G Rabchevsky
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536; Department of Physiology, University of Kentucky, Lexington, KY 40536
| | - Patrick G Sullivan
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536; Department of Anatomy and Neurobiology, University of Kentucky, Lexington, KY 40536.
| |
Collapse
|
64
|
Alwis DS, Johnstone V, Yan E, Rajan R. Diffuse traumatic brain injury and the sensory brain. Clin Exp Pharmacol Physiol 2014; 40:473-83. [PMID: 23611812 DOI: 10.1111/1440-1681.12100] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 04/17/2013] [Indexed: 01/06/2023]
Abstract
In this review we discuss the consequences to the brain's cortex, specifically to the sensory cortex, of traumatic brain injury. The thesis underlying this approach is that long-term deficits in cognition seen after brain damage in humans are likely underpinned by an impaired cortical processing of the sensory information needed to drive cognition or to be used by cognitive processes to produce a response. We take it here that the impairment to sensory processing does not arise from damage to peripheral sensory systems, but from disordered brain processing of sensory input.
Collapse
Affiliation(s)
- Dasuni S Alwis
- Department of Physiology, Monash University, Melbourne, Vic., Australia
| | | | | | | |
Collapse
|
65
|
Gu YL, Zhang LW, Ma N, Ye LL, Wang DX, Gao X. Cognitive improvement of mice induced by exercise prior to traumatic brain injury is associated with cytochrome c oxidase. Neurosci Lett 2014; 570:86-91. [PMID: 24746931 DOI: 10.1016/j.neulet.2014.04.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 04/04/2014] [Accepted: 04/05/2014] [Indexed: 12/01/2022]
Abstract
Though the evidence demonstrated that voluntary exercise programs could be implemented to enhance recovery of cognitive function induced by traumatic brain injury (TBI), the exact mechanisms were still not known. We proposed that the cognitive improvement induced by exercise in TBI mice is associated with cytochrome c oxidase (COX). To demonstrate this hypothesis, adult mice were housed with or without access to a running wheel (RW) for three weeks followed by TBI operation. Acquisition of spatial learning and memory retention was assessed by using the Morris Water Maze (MWM) on days 15 post TBI. The synaptic density was measured by Golji staining. Immunohistochemistry (IHC) for NeuN, GFAP and growth associated protein 43 (GAP43) were also performed. Using Western blot, the expressions of COX I, II, III, BDNF, synapsin I, synaptophysin (SYP) and GAP43 in hippocampus of TBI mice were determinated. Lastly, CcO activity and ATP amount were also detected. Results showed that voluntary exercise prior TBI: (i) counteracted the cognitive deficits and neuron and synaptic density loss associated with the injury; (ii) increased the levels of COX I, II, III, BDNF, synapsin I, SYP and GAP43; (iii) switched the mitochondrial CcO activity and ATP amounts. These studies demonstrated that the COX plays an important role in exercise's cognitive effects in TBI model and also provide evidence that RW training is a promise exercise for traumatically injured mice.
Collapse
Affiliation(s)
- Ying Li Gu
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Li Wei Zhang
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Ning Ma
- The Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China
| | - Lin Lin Ye
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - De Xin Wang
- Department of Neurology, Beichen Hospital of Traditional Chinese Medicine, Tianjin 300400, China
| | - Xu Gao
- The Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
66
|
Novgorodov SA, Riley CL, Yu J, Borg KT, Hannun YA, Proia RL, Kindy MS, Gudz TI. Essential roles of neutral ceramidase and sphingosine in mitochondrial dysfunction due to traumatic brain injury. J Biol Chem 2014; 289:13142-54. [PMID: 24659784 DOI: 10.1074/jbc.m113.530311] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In addition to immediate brain damage, traumatic brain injury (TBI) initiates a cascade of pathophysiological events producing secondary injury. The biochemical and cellular mechanisms that comprise secondary injury are not entirely understood. Herein, we report a substantial deregulation of cerebral sphingolipid metabolism in a mouse model of TBI. Sphingolipid profile analysis demonstrated increases in sphingomyelin species and sphingosine concurrently with up-regulation of intermediates of de novo sphingolipid biosynthesis in the brain. Investigation of intracellular sites of sphingosine accumulation revealed an elevation of sphingosine in mitochondria due to the activation of neutral ceramidase (NCDase) and the reduced activity of sphingosine kinase 2 (SphK2). The lack of change in gene expression suggested that post-translational mechanisms are responsible for the shift in the activities of both enzymes. Immunoprecipitation studies revealed that SphK2 is complexed with NCDase and cytochrome oxidase (COX) subunit 1 in mitochondria and that brain injury hindered SphK2 association with the complex. Functional studies showed that sphingosine accumulation resulted in a decreased activity of COX, a rate-limiting enzyme of the mitochondrial electron transport chain. Knocking down NCDase reduced sphingosine accumulation in mitochondria and preserved COX activity after the brain injury. Also, NCDase knockdown improved brain function recovery and lessened brain contusion volume after trauma. These studies highlight a novel mechanism of secondary TBI involving a disturbance of sphingolipid-metabolizing enzymes in mitochondria and suggest a critical role for mitochondrial sphingosine in promoting brain injury after trauma.
Collapse
Affiliation(s)
- Sergei A Novgorodov
- From the Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29401
| | | | | | | | | | | | | | | |
Collapse
|
67
|
Prieto DA, Ye X, Veenstra TD. Proteomic analysis of traumatic brain injury: the search for biomarkers. Expert Rev Proteomics 2014; 5:283-91. [DOI: 10.1586/14789450.5.2.283] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
68
|
Xing G, Barry ES, Benford B, Grunberg NE, Li H, Watson WD, Sharma P. Impact of repeated stress on traumatic brain injury-induced mitochondrial electron transport chain expression and behavioral responses in rats. Front Neurol 2013; 4:196. [PMID: 24376434 PMCID: PMC3859919 DOI: 10.3389/fneur.2013.00196] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 11/19/2013] [Indexed: 12/31/2022] Open
Abstract
A significant proportion of the military personnel returning from Iraq and Afghanistan conflicts have suffered from both mild traumatic brain injury (mTBI) and post-traumatic stress disorder. The mechanisms are unknown. We used a rat model of repeated stress and mTBI to examine brain activity and behavioral function. Adult male Sprague-Dawley rats were divided into four groups: Naïve; 3 days repeated tail-shock stress; lateral fluid percussion mTBI; and repeated stress followed by mTBI (S-mTBI). Open field activity, sensorimotor responses, and acoustic startle responses (ASRs) were measured at various time points after mTBI. The protein expression of mitochondrial electron transport chain (ETC) complex subunits (CI-V) and pyruvate dehydrogenase (PDHE1α1) were determined in four brain regions at day 7-post mTBI. Compared to Naïves, repeated stress decreased horizontal activity; repeated stress and mTBI both decreased vertical activity; and the mTBI and S-mTBI groups were impaired in sensorimotor and ASRs. Repeated stress significantly increased CI, CII, and CIII protein levels in the prefrontal cortex (PFC), but decreased PDHE1α1 protein in the PFC and cerebellum, and decreased CIV protein in the hippocampus. The mTBI treatment decreased CV protein levels in the ipsilateral hippocampus. The S-mTBI treatment resulted in increased CII, CIII, CIV, and CV protein levels in the PFC, increased CI level in the cerebellum, and increased CIII and CV levels in the cerebral cortex, but decreased CI, CII, CIV, and PDHE1α1 protein levels in the hippocampus. Thus, repeated stress or mTBI alone differentially altered ETC expression in heterogeneous brain regions. Repeated stress followed by mTBI had synergistic effects on brain ETC expression, and resulted in more severe behavioral deficits. These results suggest that repeated stress could have contributed to the high incidence of long-term neurologic and neuropsychiatric morbidity in military personnel with or without mTBI.
Collapse
Affiliation(s)
- Guoqiang Xing
- Department of Anesthesiology, Uniformed Services University of the Health Sciences , Bethesda, MD , USA
| | - Erin S Barry
- Department of Medical and Clinical Psychology, Uniformed Services University of the Health Sciences , Bethesda, MD , USA
| | - Brandi Benford
- Department of Anesthesiology, Uniformed Services University of the Health Sciences , Bethesda, MD , USA
| | - Neil E Grunberg
- Department of Medical and Clinical Psychology, Uniformed Services University of the Health Sciences , Bethesda, MD , USA
| | - He Li
- Department of Psychiatry, Uniformed Services University of the Health Sciences , Bethesda, MD , USA
| | - William D Watson
- Department of Neurology, Uniformed Services University of the Health Sciences , Bethesda, MD , USA
| | - Pushpa Sharma
- Department of Anesthesiology, Uniformed Services University of the Health Sciences , Bethesda, MD , USA
| |
Collapse
|
69
|
ROS and brain diseases: the good, the bad, and the ugly. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:963520. [PMID: 24381719 PMCID: PMC3871919 DOI: 10.1155/2013/963520] [Citation(s) in RCA: 255] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 11/04/2013] [Accepted: 11/05/2013] [Indexed: 12/31/2022]
Abstract
The brain is a major metabolizer of oxygen and yet has relatively feeble protective antioxidant mechanisms. This paper reviews the Janus-faced properties of reactive oxygen species. It will describe the positive aspects of moderately induced ROS but it will also outline recent research findings concerning the impact of oxidative and nitrooxidative stress on neuronal structure and function in neuropsychiatric diseases, including major depression. A common denominator of all neuropsychiatric diseases including schizophrenia and ADHD is an increased inflammatory response of the brain caused either by an exposure to proinflammatory agents during development or an accumulation of degenerated neurons, oxidized proteins, glycated products, or lipid peroxidation in the adult brain. Therefore, modulation of the prooxidant-antioxidant balance provides a therapeutic option which can be used to improve neuroprotection in response to oxidative stress. We also discuss the neuroprotective role of the nuclear factor erythroid 2-related factor (Nrf2) in the aged brain in response to oxidative stressors and nanoparticle-mediated delivery of ROS-scavenging drugs. The antioxidant therapy is a novel therapeutic strategy. However, the available drugs have pleiotropic actions and are not fully characterized in the clinic. Additional clinical trials are needed to assess the risks and benefits of antioxidant therapies for neuropsychiatric disorders.
Collapse
|
70
|
Bartnik-Olson BL, Harris NG, Shijo K, Sutton RL. Insights into the metabolic response to traumatic brain injury as revealed by (13)C NMR spectroscopy. FRONTIERS IN NEUROENERGETICS 2013; 5:8. [PMID: 24109452 PMCID: PMC3790078 DOI: 10.3389/fnene.2013.00008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 09/12/2013] [Indexed: 12/11/2022]
Abstract
The present review highlights critical issues related to cerebral metabolism following traumatic brain injury (TBI) and the use of (13)C labeled substrates and nuclear magnetic resonance (NMR) spectroscopy to study these changes. First we address some pathophysiologic factors contributing to metabolic dysfunction following TBI. We then examine how (13)C NMR spectroscopy strategies have been used to investigate energy metabolism, neurotransmission, the intracellular redox state, and neuroglial compartmentation following injury. (13)C NMR spectroscopy studies of brain extracts from animal models of TBI have revealed enhanced glycolytic production of lactate, evidence of pentose phosphate pathway (PPP) activation, and alterations in neuronal and astrocyte oxidative metabolism that are dependent on injury severity. Differential incorporation of label into glutamate and glutamine from (13)C labeled glucose or acetate also suggest TBI-induced adaptations to the glutamate-glutamine cycle.
Collapse
|
71
|
Arun P, Abu-Taleb R, Oguntayo S, Wang Y, Valiyaveettil M, Long JB, Nambiar MP. Acute mitochondrial dysfunction after blast exposure: potential role of mitochondrial glutamate oxaloacetate transaminase. J Neurotrauma 2013; 30:1645-51. [PMID: 23600763 DOI: 10.1089/neu.2012.2834] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Use of improvised explosive devices has significantly increased the incidence of traumatic brain injury (TBI) and associated neuropsychiatric deficits in the recent wars in Iraq and Afghanistan. Acute deleterious effects of single and repeated blast exposure can lead to long-term neurobiological effects and neuropsychiatric deficits. Using in vitro and in vivo shock tube models of blast-induced TBI, we studied changes in mitochondrial energy metabolism after blast exposure. Single and repeated blast exposures in vitro resulted in significant decreases in neuronal adenosine triphosphate (ATP) levels at 6 h post-blast that returned towards normal levels by 24 h. Similar changes in ATP also were observed in the cerebral cortices of mice subjected to single and repeated blast exposures. In neurons, mitochondrial glutamate oxaloacetate transaminase (GOT2) plays a critical role in metabolism and energy production. Proteomic analysis of brain cortices showed a significant decrease in GOT2 levels 6 h after repeated blast exposures, which was further confirmed by Western blotting. Western blot analysis of GOT2 and pyruvate dehydrogenase in the cortex showed direct correlation only between GOT2 and ATP levels. Activity of GOT2 in the isolated cortical mitochondria also showed significant decrease at 6 h supporting the results of proteomic and Western blot analyses. Knowing the significant role of GOT2 in the neuronal mitochondrial energy metabolism, it is quite likely that the down regulation of GOT2 after blast exposure is playing a significant role in mitochondrial dysfunction after blast exposure.
Collapse
Affiliation(s)
- Peethambaran Arun
- Blast-Induced Neurotrauma Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | | | | | | | | | | | | |
Collapse
|
72
|
Silva LFA, Hoffmann MS, Gerbatin RDR, Fiorin FDS, Dobrachinski F, Mota BC, Wouters ATB, Pavarini SP, Soares FAA, Fighera MR, Royes LFF. Treadmill exercise protects against pentylenetetrazol-induced seizures and oxidative stress after traumatic brain injury. J Neurotrauma 2013; 30:1278-87. [PMID: 23530735 PMCID: PMC3713448 DOI: 10.1089/neu.2012.2577] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Traumatic brain injury (TBI) is a major cause of acquired epilepsy, and significant resources are required to develop a better understanding of the pathologic mechanism as targets for potential therapies. Thus, we decided to investigate whether physical exercise after fluid percussion injury (FPI) protects from oxidative and neurochemical alterations as well as from behavioral electroencephalographic (EEG) seizures induced by subeffective convulsive doses of pentylenetetrazol (PTZ; 35 mg/kg). Behavioral and EEG recordings revealed that treadmill physical training increased latency to first clonic and tonic-clonic seizures, attenuated the duration of generalized seizures, and protected against the increase of PTZ-induced Racine scale 5 weeks after neuronal injury. EEG recordings also revealed that physical exercise prevented PTZ-induced amplitude increase in TBI animals. Neurochemical analysis showed that exercise training increased glutathione/oxidized glutathione ratio and glutathione levels per se. Exercise training was also effective against alterations in the redox status, herein characterized by lipid peroxidation (thiobarbituric acid reactive substances), protein carbonyl increase, as well as the inhibition of superoxide dismutase and Na⁺,K⁺-ATPase activities after FPI. On the other hand, histologic analysis with hematoxylin and eosin revealed that FPI induced moderate neuronal damage in cerebral cortex 4 weeks after injury and that physical exercise did not protect against neuronal injury. These data suggest that the ability of physical exercise to reduce FPI-induced seizures is not related to its protection against neuronal damage; however, the effective protection of selected targets, such as Na⁺/K⁺-ATPase elicited by physical exercise, may represent a new line of treatment for post-traumatic seizure susceptibility.
Collapse
Affiliation(s)
- Luiz Fernando Almeida Silva
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Departamento de Química, Universidade Federal de Santa Maria, 97105-900 Santa Maria, Rio Grande do Sul, Brasil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Jha MK, Jeon S, Suk K. Pyruvate Dehydrogenase Kinases in the Nervous System: Their Principal Functions in Neuronal-glial Metabolic Interaction and Neuro-metabolic Disorders. Curr Neuropharmacol 2013; 10:393-403. [PMID: 23730261 PMCID: PMC3520047 DOI: 10.2174/157015912804143586] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 08/10/2012] [Accepted: 08/28/2012] [Indexed: 01/03/2023] Open
Abstract
Metabolism is involved directly or indirectly in all processes conducted in living cells. The brain, popularly viewed as a neuronal-glial complex, gets most of its energy from the oxygen-dependent metabolism of glucose, and the mitochondrial pyruvate dehydrogenase complex (PDC) plays a key regulatory role during the oxidation of glucose. Pyruvate dehydrogenase kinase (also called PDC kinase or PDK) is a kinase that regulates glucose metabolism by switching off PDC. Four isoforms of PDKs with tissue specific activities have been identified. The metabolisms of neurons and glial cells, especially, those of astroglial cells, are interrelated, and these cells function in an integrated fashion. The energetic coupling between neuronal and astroglial cells is essential to meet the energy requirements of the brain in an efficient way. Accumulating evidence suggests that alterations in the PDKs and/or neuron-astroglia metabolic interactions are associated with the development of several neurological disorders. Here, the authors review the results of recent research efforts that have shed light on the functions of PDKs in the nervous system, particularly on neuron-glia metabolic interactions and neuro-metabolic disorders.
Collapse
Affiliation(s)
- Mithilesh Kumar Jha
- Department of Pharmacology, Brain Science & Engineering Institute, Kyungpook National University School of Medicine, Daegu, Korea
| | | | | |
Collapse
|
74
|
Guingab-Cagmat JD, Cagmat EB, Hayes RL, Anagli J. Integration of proteomics, bioinformatics, and systems biology in traumatic brain injury biomarker discovery. Front Neurol 2013; 4:61. [PMID: 23750150 PMCID: PMC3668328 DOI: 10.3389/fneur.2013.00061] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 05/12/2013] [Indexed: 01/18/2023] Open
Abstract
Traumatic brain injury (TBI) is a major medical crisis without any FDA-approved pharmacological therapies that have been demonstrated to improve functional outcomes. It has been argued that discovery of disease-relevant biomarkers might help to guide successful clinical trials for TBI. Major advances in mass spectrometry (MS) have revolutionized the field of proteomic biomarker discovery and facilitated the identification of several candidate markers that are being further evaluated for their efficacy as TBI biomarkers. However, several hurdles have to be overcome even during the discovery phase which is only the first step in the long process of biomarker development. The high-throughput nature of MS-based proteomic experiments generates a massive amount of mass spectral data presenting great challenges in downstream interpretation. Currently, different bioinformatics platforms are available for functional analysis and data mining of MS-generated proteomic data. These tools provide a way to convert data sets to biologically interpretable results and functional outcomes. A strategy that has promise in advancing biomarker development involves the triad of proteomics, bioinformatics, and systems biology. In this review, a brief overview of how bioinformatics and systems biology tools analyze, transform, and interpret complex MS datasets into biologically relevant results is discussed. In addition, challenges and limitations of proteomics, bioinformatics, and systems biology in TBI biomarker discovery are presented. A brief survey of researches that utilized these three overlapping disciplines in TBI biomarker discovery is also presented. Finally, examples of TBI biomarkers and their applications are discussed.
Collapse
|
75
|
Cortes DF, Landis MK, Ottens AK. High-capacity peptide-centric platform to decode the proteomic response to brain injury. Electrophoresis 2012; 33:3712-9. [PMID: 23160985 DOI: 10.1002/elps.201200341] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 09/12/2012] [Accepted: 09/20/2012] [Indexed: 02/05/2023]
Abstract
Traumatic brain injury (TBI) is a progressive disease process underlain by dynamic and interactive biochemical mechanisms; thus, large-scale and unbiased assessments are needed to fully understand its highly complex pathobiology. Here, we report on a new high-capacity label-free proteomic platform to evaluate the post-TBI neuroproteome. Six orthogonal separation stages and data-independent MS were employed, affording reproducible quantitative assessment on 18 651 peptides across biological replicates. From these data 3587 peptides were statistically responsive to TBI of which 18% were post-translationally modified. Results revealed as many as 484 proteins in the post-TBI neuroproteome, which was fully nine times the number determined from our prior study of focal cortical injury. Yet, these data were generated using 25 times less brain tissue per animal relative to former methodology, permitting greater anatomical specificity and proper biological replication for increased statistical power. Exemplified by these data, we discuss benefits of peptide-centric differential analysis to more accurately infer novel biological findings testable in future hypothesis-driven research. The high-capacity label-free proteomic platform is designed for multi-factor studies aimed at expanding our knowledge on the molecular underpinnings of TBI and to develop better diagnostics and therapeutics.
Collapse
Affiliation(s)
- Diego F Cortes
- Department of Anatomy & Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | | | | |
Collapse
|
76
|
Porter KM, Sutliff RL. HIV-1, reactive oxygen species, and vascular complications. Free Radic Biol Med 2012; 53:143-59. [PMID: 22564529 PMCID: PMC3377788 DOI: 10.1016/j.freeradbiomed.2012.03.019] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 03/16/2012] [Accepted: 03/18/2012] [Indexed: 02/07/2023]
Abstract
Over 1 million people in the United States and 33 million individuals worldwide suffer from HIV/AIDS. Since its discovery, HIV/AIDS has been associated with an increased susceptibility to opportunistic infection due to immune dysfunction. Highly active antiretroviral therapies restore immune function and, as a result, people infected with HIV-1 are living longer. This improved survival of HIV-1 patients has revealed a previously unrecognized risk of developing vascular complications, such as atherosclerosis and pulmonary hypertension. The mechanisms underlying these HIV-associated vascular disorders are poorly understood. However, HIV-induced elevations in reactive oxygen species (ROS), including superoxide and hydrogen peroxide, may contribute to vascular disease development and progression by altering cell function and redox-sensitive signaling pathways. In this review, we summarize the clinical and experimental evidence demonstrating HIV- and HIV antiretroviral therapy-induced alterations in reactive oxygen species and how these effects are likely to contribute to vascular dysfunction and disease.
Collapse
Affiliation(s)
- Kristi M Porter
- Pulmonary, Allergy and Critical Care Division, Emory University School of Medicine/Atlanta VA Medical Center, 1670 Clairmont Road, Mailstop 151P, Decatur, GA 30033, USA.
| | | |
Collapse
|
77
|
Sabens Liedhegner EA, Gao XH, Mieyal JJ. Mechanisms of altered redox regulation in neurodegenerative diseases--focus on S--glutathionylation. Antioxid Redox Signal 2012; 16:543-66. [PMID: 22066468 PMCID: PMC3270051 DOI: 10.1089/ars.2011.4119] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE Neurodegenerative diseases are characterized by progressive loss of neurons. A common feature is oxidative stress, which arises when reactive oxygen species (ROS) and/or reactive nitrogen species (RNS) exceed amounts required for normal redox signaling. An imbalance in ROS/RNS alters functionality of cysteines and perturbs thiol-disulfide homeostasis. Many cysteine modifications may occur, but reversible protein mixed disulfides with glutathione (GSH) likely represents the common steady-state derivative due to cellular abundance of GSH and ready conversion of cysteine-sulfenic acid and S-nitrosocysteine precursors to S-glutathionylcysteine disulfides. Thus, S-glutathionylation acts in redox signal transduction and serves as a protective mechanism against irreversible cysteine oxidation. Reversal of protein-S-glutathionylation is catalyzed specifically by glutaredoxin which thereby plays a critical role in cellular regulation. This review highlights the role of oxidative modification of proteins, notably S-glutathionylation, and alterations in thiol homeostatic enzyme activities in neurodegenerative diseases, providing insights for therapeutic intervention. RECENT ADVANCES Recent studies show that dysregulation of redox signaling and sulfhydryl homeostasis likely contributes to onset/progression of neurodegeneration. Oxidative stress alters the thiol-disulfide status of key proteins that regulate the balance between cell survival and cell death. CRITICAL ISSUES Much of the current information about redox modification of key enzymes and signaling intermediates has been gleaned from studies focused on oxidative stress situations other than the neurodegenerative diseases. FUTURE DIRECTIONS The findings in other contexts are expected to apply to understanding neurodegenerative mechanisms. Identification of selectively glutathionylated proteins in a quantitative fashion will provide new insights about neuropathological consequences of this oxidative protein modification.
Collapse
|
78
|
Patel SP, Sullivan PG, Lyttle TS, Magnuson DSK, Rabchevsky AG. Acetyl-L-carnitine treatment following spinal cord injury improves mitochondrial function correlated with remarkable tissue sparing and functional recovery. Neuroscience 2012; 210:296-307. [PMID: 22445934 DOI: 10.1016/j.neuroscience.2012.03.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Revised: 02/22/2012] [Accepted: 03/02/2012] [Indexed: 01/10/2023]
Abstract
We have recently documented that treatment with the alternative biofuel, acetyl-L-carnitine (ALC, 300 mg/kg), as late as 1 h after T10 contusion spinal cord injury (SCI), significantly maintained mitochondrial function 24 h after injury. Here we report that after more severe contusion SCI centered on the L1/L2 segments that are postulated to contain lamina X neurons critical for locomotion (the "central pattern generator"), ALC treatment resulted in significant improvements in acute mitochondrial bioenergetics and long-term hind limb function. Although control-injured rats were only able to achieve slight movements of hind limb joints, ALC-treated animals produced consistent weight-supported plantar steps 1 month after injury. Such landmark behavioral improvements were significantly correlated with increased tissue sparing of both gray and white matter proximal to the injury, as well as preservation of choline acetyltransferase (ChAT)-positive neurons in lamina X rostral to the injury site. These findings signify that functional improvements with ALC treatment are mediated, in part, by preserved locomotor circuitry rostral to upper lumbar contusion SCI. Based on beneficial effects of ALC on mitochondrial bioenergetics after injury, our collective evidence demonstrate that preventing mitochondrial dysfunction acutely "promotes" neuroprotection that may be associated with the milestone recovery of plantar, weight-supported stepping.
Collapse
Affiliation(s)
- S P Patel
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536-0509, USA
| | | | | | | | | |
Collapse
|
79
|
Xing G, Ren M, O'Neill JT, Verma A, Watson WD. Controlled cortical impact injury and craniotomy result in divergent alterations of pyruvate metabolizing enzymes in rat brain. Exp Neurol 2012; 234:31-38. [PMID: 22193111 DOI: 10.1016/j.expneurol.2011.12.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 12/04/2011] [Indexed: 01/04/2023]
Abstract
Dysregulated glucose metabolism and energy deficit is a characteristic of severe traumatic brain injury (TBI) but its mechanism remains to be fully elucidated. Phosphorylation of pyruvate dehydrogenase (PDH) is the rate-limiting mitochondria enzyme reaction coupling glycolysis to the tricarboxylic acid cycle. Phosphorylation of PDH E1α1 subunit catalyzed by PDH kinase (PDK) inhibits PDH activity, effectively decoupling aerobic glycolysis whereas dephosphorylation of phosphorylated PDHE1α1 by PDH phosphatase (PDP) restores PDH activity. We recently reported altered expression and phosphorylation of pyruvate dehydrogenase (PDH) following TBI. However, little is known about PDK and PDP involvement. We determined PDK (PDK1-4) and PDP isoenzyme (PDP1-2) mRNA and protein expression in rat brain using immunohistochemistry and in situ hybridization techniques. We also quantified PDK and PDP mRNA and protein levels in rat brain following TBI using quantitative real-time PCR and Western blot, respectively. Controlled cortical impact-induced TBI (CCI-TBI) and craniotomy significantly enhanced PDK1-2 isoenzyme mRNA expression level but significantly suppressed PDP1 and PDK4 mRNA expression after the injury (4h to 7days). CCI-TBI and craniotomy also significantly increased PDK1-4 isoenzyme protein expression but suppressed PDP1-2 protein expression in rat brain. In summary, the divergent changes between PDK and PDP expression indicate imbalance between PDK and PDP activities that would favor increased PDHE1α1 phosphorylation and enzyme inhibition contributing to impaired oxidative glucose metabolism in TBI as well as craniotomy.
Collapse
Affiliation(s)
- Guoqiang Xing
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814-4799, USA.
| | | | | | | | | |
Collapse
|
80
|
Saraiva ALL, Ferreira APO, Silva LFA, Hoffmann MS, Dutra FD, Furian AF, Oliveira MS, Fighera MR, Royes LFF. Creatine reduces oxidative stress markers but does not protect against seizure susceptibility after severe traumatic brain injury. Brain Res Bull 2011; 87:180-6. [PMID: 22051612 DOI: 10.1016/j.brainresbull.2011.10.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2011] [Revised: 10/04/2011] [Accepted: 10/20/2011] [Indexed: 12/22/2022]
Abstract
Achievements made over the last years have highlighted the important role of creatine in health and disease. However, its effects on hyperexcitable circuit and oxidative damage induced by traumatic brain injury (TBI) are not well understood. In the present study we revealed that severe TBI elicited by fluid percussion brain injury induced oxidative damage characterized by protein carbonylation, thiobarbituric acid reactive species (TBARS) increase and Na(+),K(+)-ATPase activity inhibition 4 and 8 days after neuronal injury. Statistical analysis showed that after TBI creatine supplementation (300 mg/kg, p.o.) decreased the levels of protein carbonyl and TBARS but did not protect against TBI-induced Na(+),K(+)-ATPase activity inhibition. Electroencephalography (EEG) analysis revealed that the injection of a subconvulsant dose of PTZ (35 mg/kg, i.p.), 4 but not 8 days after neuronal injury, decreased latency for the first clonic seizures and increased the time of spent generalized tonic-clonic seizures compared with the sham group. In addition, creatine supplementation had no effect on convulsive parameters induced by a subconvulsant dose of PTZ. Current experiments provide evidence that lipid and protein oxidation represents a separate pathway in the early post-traumatic seizures susceptibility. Furthermore, the lack of consistent anticonvulsant effect exerted by creatine in this early phase suggests that its apparent antioxidant effect does not protect against excitatory input generation induced by TBI.
Collapse
Affiliation(s)
- André Luis Lopes Saraiva
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Sultana R, Robinson RAS, Di Domenico F, Mohmmad Abdul H, St. Clair DK, Markesbery WR, Cai J, Pierce WM, Butterfield DA. Proteomic identification of specifically carbonylated brain proteins in APP(NLh)/APP(NLh) × PS-1(P264L)/PS-1(P264L) human double mutant knock-in mice model of Alzheimer disease as a function of age. J Proteomics 2011; 74:2430-40. [PMID: 21726674 PMCID: PMC3199338 DOI: 10.1016/j.jprot.2011.06.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 06/10/2011] [Accepted: 06/15/2011] [Indexed: 10/18/2022]
Abstract
Alzheimer disease (AD) is the most common type of dementia and is characterized pathologically by the presence of neurofibrillary tangles (NFTs), senile plaques (SPs), and loss of synapses. The main component of SP is amyloid-beta peptide (Aβ), a 39 to 43 amino acid peptide, generated by the proteolytic cleavage of amyloid precursor protein (APP) by the action of beta- and gamma-secretases. The presenilins (PS) are components of the γ-secretase, which contains the protease active center. Mutations in PS enhance the production of the Aβ42 peptide. To date, more than 160 mutations in PS1 have been identified. Many PS mutations increase the production of the β-secretase-mediated C-terminal (CT) 99 amino acid-long fragment (CT99), which is subsequently cleaved by γ-secretase to yield Aβ peptides. Aβ has been proposed to induce oxidative stress and neurotoxicity. Previous studies from our laboratory and others showed an age-dependent increase in oxidative stress markers, loss of lipid asymmetry, and Aβ production and amyloid deposition in the brain of APP/PS1 mice. In the present study, we used APP (NLh)/APP(NLh) × PS-1(P246L)/PS-1(P246L) human double mutant knock-in APP/PS-1 mice to identify specific targets of brain protein carbonylation in an age-dependent manner. We found a number of proteins that are oxidatively modified in APP/PS1 mice compared to age-matched controls. The relevance of the identified proteins to the progression and pathogenesis of AD is discussed.
Collapse
Affiliation(s)
- Rukhsana Sultana
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506
| | - Renã A. S. Robinson
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506
| | - Fabio Di Domenico
- Department of Biochemical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Hafiz Mohmmad Abdul
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506
| | - Daret K. St. Clair
- Graduate Center of Toxicology, University of Kentucky, Lexington, KY 40536, USA
| | | | - Jian Cai
- Department of Pharmacology, University of Louisville, Louisville, Kentucky 40292
| | - William M. Pierce
- Department of Pharmacology, University of Louisville, Louisville, Kentucky 40292
| | - D. Allan Butterfield
- Department of Chemistry, Center of Membrane Sciences, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506
| |
Collapse
|
82
|
Mechanisms for endothelial monocyte-activating polypeptide-II-induced opening of the blood-tumor barrier. J Mol Neurosci 2011; 47:408-17. [PMID: 21969114 DOI: 10.1007/s12031-011-9657-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 09/22/2011] [Indexed: 10/17/2022]
Abstract
Endothelial monocyte-activating polypeptide-II (EMAP-II) increases blood-tumor barrier (BTB) permeability by inducing alterations in the tight junction (TJ) complex between brain endothelial cells. In the present study, an in vitro BTB model was used to search for the interacting and functional cell surface molecule of EMAP-II as well as the signaling pathway involved in the EMAP-II-induced BTB hyperpermeability. Our results revealed that EMAP-II-induced increase in BTB permeability and down-regulation of TJ-related proteins occludin and ZO-1 were associated with its binding to ATP synthase α subunit (α-ATP synthase) on the surface of rat brain microvascular endothelial cells (BMECs). In addition, we observed that EMAP-II administration activated protein kinase C (PKC) and induced the translocation of PKC from the cytosolic to the membrane fraction of BMECs. The effects of EMAP-II on BTB permeability as well as expression levels of occludin and ZO-1 in BMECs were significantly diminished by H7, the inhibitor of PKC. In summary, these data suggest that EMAP-II increases BTB permeability through α-ATP synthase on the surface of BMECs, and PKC signaling pathway might be involved in this process.
Collapse
|
83
|
Readnower RD, Pandya JD, McEwen ML, Pauly JR, Springer JE, Sullivan PG. Post-injury administration of the mitochondrial permeability transition pore inhibitor, NIM811, is neuroprotective and improves cognition after traumatic brain injury in rats. J Neurotrauma 2011; 28:1845-53. [PMID: 21875332 DOI: 10.1089/neu.2011.1755] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mitochondrial dysfunction is known to play a pivotal role in cell death mechanisms following traumatic brain injury (TBI). N-methyl-4-isoleucine-cyclosporin (NIM811), a non-immunosuppressive cyclosporin A (CsA) analog, inhibits the mitochondrial permeability transition pore (mPTP) and has been shown to be neuroprotective following TBI in mice. However, the translation of the neuroprotective effects of mPTP inhibitors, including CsA and NIM811, into improved cognitive end points has yet to be fully investigated. Therefore, to build upon these results, a severe unilateral controlled cortical impact model of TBI was used in the present study to establish a dose-response curve for NIM811 in rats. The findings demonstrate that the neuroprotection afforded by NIM811 is dose dependent, with the 10 mg/kg dose being the most effective dose. Once the dose response was established, we evaluated the effect of the optimal dose of NIM811 on behavior, mitochondrial bioenergetics, and mitochondrial oxidative damage following TBI. For behavioral studies, rats were administered NIM811 at 15 min and 24 h post-injury, with cognitive testing beginning 10 days post-injury. Mitochondrial studies involved a single injection of NIM811 at 15 min post-injury followed by mitochondrial isolation at 6 h post-injury. The results revealed that the optimal dose of NIM811 improves cognition, improves mitochondrial functioning, and reduces oxidative damage following TBI.
Collapse
Affiliation(s)
- Ryan D Readnower
- Spinal Cord & Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | | | | | | | | | | |
Collapse
|
84
|
Deng-Bryant Y, Prins ML, Hovda DA, Harris NG. Ketogenic diet prevents alterations in brain metabolism in young but not adult rats after traumatic brain injury. J Neurotrauma 2011; 28:1813-25. [PMID: 21635175 DOI: 10.1089/neu.2011.1822] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Previous studies have shown that the change of cerebral metabolic rate of glucose (CMRglc) in response to traumatic brain injury (TBI) is different in young (PND35) and adult rats (PND70), and that prolonged ketogenic diet treatment results in histological and behavioral neuroprotection only in younger rat brains. However, the mechanism(s) through which ketones act in the injured brain and the biochemical markers of their action remain unknown. Therefore, the current study was initiated to: 1) determine the effect of injury on the neurochemical profile in PND35 compared to PND70 rats; and 2) test the effect of early post-injury administration of ketogenic diet on brain metabolism in PND35 versus PND70 rats. The data show that alterations in energy metabolites, amino acid, and membrane metabolites were not evident in PND35 rats on standard diet until 24 h after injury, when the concentration of most metabolites was reduced from sham-injured values. In contrast, acute, but transient deficits in energy metabolism were measured at 6 h in PND70 rats, together with deficits in N-acetylaspartate that endured until 24 h. Administration of a ketogenic diet resulted in significant increases in plasma β-hydroxybutyrate (βOHB) levels. Similarly, brain βOHB levels were significantly elevated in all injured rats, but were elevated by 43% more in PND35 rats compared to PND70 rats. As a result, ATP, creatine, and phosphocreatine levels at 24 h after injury were significantly improved in the ketogenic PND35 rats, but not in the PND70 group. The improvement in energy metabolism in the PND35 brains was accompanied by the recovery of NAA and reduction of lactate levels, as well as amelioration of the deficits of other amino acids and membrane metabolites. These results indicate that the PND35 brains are more resistant to the injury, indicated by a delayed deficit in energy metabolism. Moreover, the younger brains revert to ketones metabolism more quickly than do the adult brains, resulting in better neurochemical and cerebral metabolic recovery after injury.
Collapse
Affiliation(s)
- Ying Deng-Bryant
- Department of Neurosurgery, UCLA David Geffen School of Medicine, Los Angeles, California, USA.
| | | | | | | |
Collapse
|
85
|
Rabchevsky AG, Patel SP, Springer JE. Pharmacological interventions for spinal cord injury: where do we stand? How might we step forward? Pharmacol Ther 2011; 132:15-29. [PMID: 21605594 DOI: 10.1016/j.pharmthera.2011.05.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 04/28/2011] [Indexed: 12/15/2022]
Abstract
Despite numerous studies reporting some measures of efficacy in the animal literature, there are currently no effective therapies for the treatment of traumatic spinal cord injuries (SCI) in humans. The purpose of this review is to delineate key pathophysiological processes that contribute to neurological deficits after SCI, as well as to describe examples of pharmacological approaches that are currently being tested in clinical trials, or nearing clinical translation, for the therapeutic management of SCI. In particular, we will describe the mechanistic rationale to promote neuroprotection and/or functional recovery based on theoretical, yet targeted pathological events. Finally, we will consider the clinical relevancy for emerging evidence that pharmacologically targeting mitochondrial dysfunction following injury may hold the greatest potential for increasing tissue sparing and, consequently, the extent of functional recovery following traumatic SCI.
Collapse
Affiliation(s)
- Alexander G Rabchevsky
- Spinal Cord & Brain injury Research Center, Lexington, University of Kentucky, KY 40536-0509, USA.
| | | | | |
Collapse
|
86
|
Oxidative Stress Parameters in Different Brain Structures Following Lateral Fluid Percussion Injury in the Rat. Neurochem Res 2011; 36:913-21. [DOI: 10.1007/s11064-011-0424-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2011] [Indexed: 02/02/2023]
|
87
|
Timofeev I, Carpenter KLH, Nortje J, Al-Rawi PG, O'Connell MT, Czosnyka M, Smielewski P, Pickard JD, Menon DK, Kirkpatrick PJ, Gupta AK, Hutchinson PJ. Cerebral extracellular chemistry and outcome following traumatic brain injury: a microdialysis study of 223 patients. Brain 2011; 134:484-94. [PMID: 21247930 DOI: 10.1093/brain/awq353] [Citation(s) in RCA: 263] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Secondary insults can adversely influence outcome following severe traumatic brain injury. Monitoring of cerebral extracellular chemistry with microdialysis has the potential for early detection of metabolic derangements associated with such events. The objective of this study was to determine the relationship between the fundamental biochemical markers and neurological outcome in a large cohort of patients with traumatic brain injury. Prospectively collected observational neuromonitoring data from 223 patients were analysed. Monitoring modalities included digitally recorded intracranial pressure, cerebral perfusion pressure, cerebrovascular pressure reactivity index and microdialysis markers glucose, lactate, pyruvate, glutamate, glycerol and the lactate/pyruvate ratio. Outcome was assessed using the Glasgow Outcome Scale at 6 months post-injury. Patient-averaged values of parameters were used in statistical analysis, which included univariate non-parametric methods and multivariate logistic regression. Monitoring with microdialysis commenced on median (interquartile range) Day 1 (1-2) from injury and median (interquartile range) duration of monitoring was 4 (2-7) days. Averaged over the total monitoring period levels of glutamate (P = 0.048), lactate/pyruvate ratio (P = 0.044), intracranial pressure (P = 0.006) and cerebrovascular pressure reactivity index (P = 0.01) were significantly higher in patients who died. During the initial 72 h of monitoring, median glycerol levels were also higher in the mortality group (P = 0.014) and median lactate/pyruvate ratio (P = 0.026) and lactate (P = 0.033) levels were significantly lower in patients with favourable outcome. In a multivariate logistic regression model (P < 0.0001), which employed data averaged over the whole monitoring period, significant independent positive predictors of mortality were glucose (P = 0.024), lactate/pyruvate ratio (P = 0.016), intracranial pressure (P = 0.029), cerebrovascular pressure reactivity index (P = 0.036) and age (P = 0.003), while pyruvate was a significant independent negative predictor of mortality (P = 0.004). The results of this study suggest that extracellular metabolic markers are independently associated with outcome following traumatic brain injury. Whether treatment-related improvement in biochemistry translates into better outcome remains to be established.
Collapse
Affiliation(s)
- Ivan Timofeev
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Addenbrooke’s Hospital, Cambridge, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Kochanek PM, Bell MJ, Bayır H. Quo vadis 2010? - carpe diem: challenges and opportunities in pediatric traumatic brain injury. Dev Neurosci 2011; 32:335-42. [PMID: 21252553 PMCID: PMC3215241 DOI: 10.1159/000323016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Accepted: 08/20/2010] [Indexed: 01/05/2023] Open
Abstract
Traumatic brain injury (TBI) in infants and children remains a public health problem of enormous magnitude. It is a complex and heterogeneous condition that presents many diagnostic, therapeutic and prognostic challenges. A number of investigative teams are studying pediatric TBI both in experimental models and in clinical studies at the bedside. This review builds on work presented in a prior supplement to Developmental Neuroscience that was published in 2006, and addresses several active areas of research on this topic, including (1) the application of novel imaging methods, (2) the use of serum and/or CSF biomarkers of injury, (3) advances in neuromonitoring, (4) the development and testing of novel therapies, (5) developments in modeling pediatric TBI, (6) the consideration of a new approach to classification of pediatric TBI, and (7) assessing the potential impact of the development of pediatric and neonatal neurocritical care services on the management and outcome of pediatric TBI.
Collapse
Affiliation(s)
- Patrick M Kochanek
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa., USA.
| | | | | |
Collapse
|
89
|
Scafidi S, Racz J, Hazelton J, McKenna MC, Fiskum G. Neuroprotection by acetyl-L-carnitine after traumatic injury to the immature rat brain. Dev Neurosci 2011; 32:480-7. [PMID: 21228558 DOI: 10.1159/000323178] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Accepted: 11/25/2010] [Indexed: 01/05/2023] Open
Abstract
Traumatic brain injury (TBI) is the leading cause of mortality and morbidity in children and is characterized by reduced aerobic cerebral energy metabolism early after injury, possibly due to impaired activity of the pyruvate dehydrogenase complex. Exogenous acetyl-L-carnitine (ALCAR) is metabolized in the brain to acetyl coenzyme A and subsequently enters the tricarboxylic acid cycle. ALCAR administration is neuroprotective in animal models of cerebral ischemia and spinal cord injury, but has not been tested for TBI. This study tested the hypothesis that treatment with ALCAR during the first 24 h following TBI in immature rats improves neurologic outcome and reduces cortical lesion volume. Postnatal day 21-22 male rats were isoflurane anesthetized and used in a controlled cortical impact model of TBI to the left parietal cortex. At 1, 4, 12 and 23 h after injury, rats received ALCAR (100 mg/kg, intraperitoneally) or drug vehicle (normal saline). On days 3-7 after surgery, behavior was assessed using beam walking and novel object recognition tests. On day 7, rats were transcardially perfused and brains were harvested for histological assessment of cortical lesion volume, using stereology. Injured animals displayed a significant increase in foot slips compared to sham-operated rats (6 ± 1 SEM vs. 2 ± 0.2 on day 3 after trauma; n = 7; p < 0.05). The ALCAR-treated rats were not different from shams and had fewer foot slips compared to vehicle-treated animals (2 ± 0.4; n = 7; p< 0.05). The frequency of investigating a novel object for saline-treated TBI animals was reduced compared to shams (45 ± 5% vs. 65 ± 10%; n = 7; p < 0.05), whereas the frequency of investigation for TBI rats treated with ALCAR was not significantly different from that of shams but significantly higher than that of saline-treated TBI rats (68 ± 7; p < 0.05). The left parietal cortical lesion volume, expressed as a percentage of the volume of tissue in the right hemisphere, was significantly smaller in ALCAR-treated than in vehicle-treated TBI rats (14 ± 5% vs. 28 ± 6%; p < 0.05). We conclude that treatment with ALCAR during the first 24 h after TBI improves behavioral outcomes and reduces brain lesion volume in immature rats within the first 7 days after injury.
Collapse
Affiliation(s)
- Susanna Scafidi
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | |
Collapse
|
90
|
Nuclear coded mitochondrial protein prohibitin is an iron regulated iron binding protein. Mitochondrion 2011; 11:40-7. [DOI: 10.1016/j.mito.2010.07.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2010] [Revised: 06/12/2010] [Accepted: 07/08/2010] [Indexed: 12/17/2022]
|
91
|
Therapeutic targets for neuroprotection and/or enhancement of functional recovery following traumatic brain injury. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 98:85-131. [PMID: 21199771 DOI: 10.1016/b978-0-12-385506-0.00003-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Traumatic brain injury (TBI) is a significant public health concern. The number of injuries that occur each year, the cost of care, and the disabilities that can lower the victim's quality of life are all driving factors for the development of therapy. However, in spite of a wealth of promising preclinical results, clinicians are still lacking a therapy. The use of preclinical models of the primary mechanical trauma have greatly advanced our knowledge of the complex biochemical sequela that follow. This cascade of molecular, cellular, and systemwide changes involves plasticity in many different neurochemical systems, which represent putative targets for remediation or attenuation of neuronal injury. The purpose of this chapter is to highlight some of the promising molecular and cellular targets that have been identified and to provide an up-to-date summary of the development of therapeutic compounds for those targets.
Collapse
|
92
|
Bartnik-Olson BL, Oyoyo U, Hovda DA, Sutton RL. Astrocyte oxidative metabolism and metabolite trafficking after fluid percussion brain injury in adult rats. J Neurotrauma 2010; 27:2191-202. [PMID: 20939699 DOI: 10.1089/neu.2010.1508] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Despite various lines of evidence pointing to the compartmentation of metabolism within the brain, few studies have reported the effect of a traumatic brain injury (TBI) on neuronal and astrocyte compartments and/or metabolic trafficking between these cells. In this study we used ex vivo ¹³C NMR spectroscopy following an infusion of [1-¹³C] glucose and [1,2-¹³C₂] acetate to study oxidative metabolism in neurons and astrocytes of sham-operated and fluid percussion brain injured (FPI) rats at 1, 5, and 14 days post-surgery. FPI resulted in a decrease in the ¹³C glucose enrichment of glutamate in neurons in the injured hemisphere at day 1. In contrast, enrichment of glutamine in astrocytes from acetate was not significantly decreased at day 1. At day 5 the ¹³C enrichment of glutamate and glutamine from glucose in the injured hemisphere of FPI rats did not differ from sham levels, but glutamine derived from acetate metabolism in astrocytes was significantly increased. The ¹³C glucose enrichment of the C3 position of glutamate (C3) in neurons was significantly decreased ipsilateral to FPI at day 14, whereas the enrichment of glutamine in astrocytes had returned to sham levels at this time point. These findings indicate that the oxidative metabolism of glucose is reduced to a greater extent in neurons compared to astrocytes following a FPI. The increased utilization of acetate to synthesize glutamine, and the acetate enrichment of glutamate via the glutamate-glutamine cycle, suggests an integral protective role for astrocytes in maintaining metabolic function following TBI-induced impairments in glucose metabolism.
Collapse
Affiliation(s)
- Brenda L Bartnik-Olson
- Brain Injury Research Center, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, California, USA.
| | | | | | | |
Collapse
|
93
|
Fukushima M, Lee SM, Moro N, Hovda DA, Sutton RL. Metabolic and histologic effects of sodium pyruvate treatment in the rat after cortical contusion injury. J Neurotrauma 2010; 26:1095-110. [PMID: 19594384 DOI: 10.1089/neu.2008.0771] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
This study determined the effects of intraperitoneal sodium pyruvate (SP) treatment on the levels of circulating fuels and on cerebral microdialysis levels of glucose (MD(glc)), lactate (MD(lac)), and pyruvate (MD(pyr)), and the effects of SP treatment on neuropathology after left cortical contusion injury (CCI) in rats. SP injection (1000 mg/kg) 5 min after sham injury (Sham-SP) or CCI (CCI-SP) significantly increased arterial pyruvate (p < 0.005) and lactate (p < 0.001) compared to that of saline-treated rats with CCI (CCI-Sal). Serum glucose also increased significantly in CCI-SP compared to that in CCI-Sal rats (p < 0.05), but not in Sham-SP rats. MD(pyr) was not altered after CCI-Sal, whereas MD(lac) levels within the cerebral cortex significantly increased bilaterally (p < 0.05) and those for MD(glc) decreased bilaterally (p < 0.05). MD(pyr) levels increased significantly in both Sham-SP and CCI-SP rats (p < 0.05 vs. CCI-Sal) and were higher in left/injured cortex of the CCI-SP group (p < 0.05 vs. sham-SP). In CCI-SP rats the contralateral MD(lac) decreased below CCI-Sal levels (p < 0.05) and the ipsilateral MD(glc) levels exceeded those of CCI-Sal rats (p < 0.05). Rats with a single low (500 mg/kg) or high dose (1000 mg/kg) SP treatment had fewer damaged cortical cells 6 h post-CCI than did saline-treated rats (p < 0.05), but three hourly injections of SP (1000 mg/kg) were needed to significantly reduce contusion volume 2 weeks after CCI. Thus, a single intraperitoneal SP treatment increases circulating levels of three potential brain fuels, attenuates a CCI-induced reduction in extracellular glucose while increasing extracellular levels of pyruvate, but not lactate, and can attenuate cortical cell damage occurring within 6 h of injury. Enduring (2 week) neuronal protection was achieved only with multiple SP treatments within the first 2 h post-CCI, perhaps reflecting the need for additional fuel throughout the acute period of increased metabolic demands induced by CCI.
Collapse
Affiliation(s)
- Masamichi Fukushima
- Brain Injury Research Center, David Geffen School of Medicine at UCLA, Los Angeles, California 90095-7039, USA
| | | | | | | | | |
Collapse
|
94
|
Gilmer LK, Roberts KN, Joy K, Sullivan PG, Scheff SW. Early mitochondrial dysfunction after cortical contusion injury. J Neurotrauma 2010; 26:1271-80. [PMID: 19637966 DOI: 10.1089/neu.2008.0857] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Following traumatic brain injury, mitochondria sustain structural and functional impairment, which contributes to secondary damage that can continue for days after the initial injury. The present study investigated mitochondrial bioenergetic changes in the rat neocortex at 1 and 3 h after mild, moderate, and severe injuries. Brains from young adult Sprague-Dawley rats were harvested from the injured and contralateral cortex to assess possible changes in mitochondrial respiration abilities following a unilateral cortical contusion injury. Differential centrifugation was used to isolate synaptic and extrasynaptic mitochondria from cortical tissue. Bioenergetics was assessed using a Clark-type electrode and results were graphed as a function of injury severity and time post-injury. Respiration was significantly affected by all injury severity levels compared to uninjured tissue. Complex 1- and complex 2-driven respirations were affected proportionally to the severity of the injury, indicating that damage to mitochondria may occur on a gradient. Total oxygen utilization, respiratory control ratio, ATP production, and maximal respiration capabilities were all significantly decreased in the injured cortex at both 1 and 3 h post-trauma. Although mitochondria displayed bioenergetic deficits at 1 h following injury, damage was not exacerbated by 3 h. This study stresses the importance of early therapeutic intervention and suggests a window of approximately 1-3 h before greater dysfunction occurs.
Collapse
Affiliation(s)
- Lesley K Gilmer
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536-0230, USA
| | | | | | | | | |
Collapse
|
95
|
Wu KLH, Hsu C, Chan JYH. Nitric oxide and superoxide anion differentially activate poly(ADP-ribose) polymerase-1 and Bax to induce nuclear translocation of apoptosis-inducing factor and mitochondrial release of cytochrome c after spinal cord injury. J Neurotrauma 2010; 26:965-77. [PMID: 19473058 DOI: 10.1089/neu.2008.0692] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
We reported previously that complete spinal cord transection (SCT) results in depression of mitochondrial respiratory chain enzyme activity that triggers apoptosis via sequential activations of apoptosis-inducing factor (AIF)- and caspase-dependent cascades in the injured spinal cord. This study tested the hypothesis that nitric oxide (NO) and superoxide anion (O(2)(.-)) serve as the interposing signals between SCT and impaired mitochondrial respiratory functions. Adult Sprague-Dawley rats manifested a significant increase in NO or O(2)(.-) level in the injured spinal cord during the first 3 days after SCT. The augmented O(2)(.-) production, along with concomitant reduction in mitochondrial respiratory chain enzyme activity or ATP level, nuclear translocation of AIF, cytosolic release of cytochrome c, and DNA fragmentation were reversed by osmotic minipump infusion of a NO trapping agent, carboxy-PTIO, or a superoxide dismutase mimetic, tempol, into the epicenter of the transected spinal cord. Intriguingly, carboxy-PTIO significantly suppressed upregulation of poly(ADP-ribose) polymerase-1 (PARP-1) in the nucleus, attenuated nuclear translocation of AIF, inhibited mitochondrial translocation of Bax and antagonized mitochondrial release of cytochrome c; whereas tempol only inhibited the later two cellular events after SCT. We conclude that overproduction of NO and O(2)(.-) in the injured spinal cord promulgates mitochondrial dysfunction and triggers AIF- and caspase-dependent apoptotic signaling cascades via differential upregulation of nuclear PARP-1 and mitochondrial translocation of Bax.
Collapse
Affiliation(s)
- Kay L H Wu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, Republic of China
| | | | | |
Collapse
|
96
|
Moro N, Sutton RL. Beneficial effects of sodium or ethyl pyruvate after traumatic brain injury in the rat. Exp Neurol 2010; 225:391-401. [PMID: 20670624 DOI: 10.1016/j.expneurol.2010.07.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Revised: 07/02/2010] [Accepted: 07/15/2010] [Indexed: 11/27/2022]
Abstract
Sodium pyruvate (SP) treatment initiated within 5 min post-injury is neuroprotective in a rat model of unilateral cortical contusion injury (CCI). The current studies examined: (1) effects of delayed SP treatments (1000 mg/kg, i.p., at 1, 12 and 24h), (2) effects of single (1h) or multiple (1, 12 and 24h) ethyl pyruvate treatments (EP; at 20 or 40 mg/kg, i.p.), and (3) mechanisms of action for pyruvate effects after CCI. In Experiment 1, both SP and EP treatment(s) significantly reduced the number of dead/dying cells in the ipsilateral hippocampus (dentate hilus+CA3(c) and/or CA3(a-b) regions) at 72 h post-CCI. Pyruvate treatment(s) attenuated CCI-induced reductions of cerebral cytochrome oxidase activity at 7 2h, significantly improving activity in peri-contusional cortex after multiple SP or EP treatments. Optical density measures of ipsilateral CD11b immuno-staining were significantly increased 72 h post-CCI, but these measures of microglia activation were not different from sham injury values in SP and EP groups with three post-CCI treatments. In Experiment 2, three treatments (1, 12 and 24h) of SP (1000 mg/kg) or EP (40 mg/kg) significantly improved recovery of beam-walking and neurological scores in the first 3 weeks after CCI, and EP treatments significantly improved spatial working memory 1 week post-CCI. Ipsilateral CA3(b) neuronal loss, but not cortical tissue loss, was significantly reduced 1 month post-CCI with pyruvate treatments begun 1h post-CCI. Thus, delayed pyruvate treatments after CCI are neuroprotective and improve neurobehavioral recovery; these effects may be mediated by improved metabolism and reduced inflammation.
Collapse
Affiliation(s)
- Nobuhiro Moro
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | | |
Collapse
|
97
|
Rosales-Corral S, Reiter RJ, Tan DX, Ortiz GG, Lopez-Armas G. Functional aspects of redox control during neuroinflammation. Antioxid Redox Signal 2010; 13:193-247. [PMID: 19951033 DOI: 10.1089/ars.2009.2629] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Neuroinflammation is a CNS reaction to injury in which some severe pathologies, regardless of their origin, converge. The phenomenon emphasizes crosstalk between neurons and glia and reveals a complex interaction with oxidizing agents through redox sensors localized in enzymes, receptors, and transcription factors. When oxidizing pressures cause reversible molecular changes, such as minimal or transitory proinflammatory cytokine overproduction, redox couples provide a means of translating the presence of reactive oxygen or nitrogen species into useful signals in the cell. Additionally, thiol-based redox sensors convey information about localized changes in redox potential induced by physiologic or pathologic situations. They are susceptible to oxidative changes and become key events during neuroinflammation, altering the course of a signaling response or the behavior of specific transcription factors. When oxidative stress augments the pressure on the intracellular environment, the effective reduction potential of redox pairs diminishes, and cell signaling shifts toward proinflammatory and proapoptotic signals, creating a vicious cycle between oxidative stress and neuroinflammation. In addition, electrophilic compounds derived from the oxidative cascade react with key protein thiols and interfere with redox signaling. This article reviews the relevant functional aspects of redox control during the neuroinflammatory process.
Collapse
Affiliation(s)
- Sergio Rosales-Corral
- Lab. Desarrollo-Envejecimiento, Enfermedades Neurodegenerativas, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO) del Instituto Mexicano del Seguro Social (IMSS) , Guadalajara, Jalisco. Mexico.
| | | | | | | | | |
Collapse
|
98
|
Roede JR, Jones DP. Reactive species and mitochondrial dysfunction: mechanistic significance of 4-hydroxynonenal. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2010; 51:380-90. [PMID: 20544880 PMCID: PMC5906392 DOI: 10.1002/em.20553] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Mitochondrial dysfunction is a global term used in the context of "unhealthy" mitochondria. In practical terms, mitochondria are extremely complex and highly adaptive in structure, chemical and enzymatic composition, subcellular distribution and functional interaction with other components of cells. Consequently, altered mitochondrial properties that are used in experimental studies as measures of mitochondrial dysfunction often provide little or no distinction between adaptive and maladaptive changes. This is especially a problem in terms of generation of oxidant species by mitochondria, wherein increased generation of superoxide anion radical (O(2*)(-)) or hydrogen peroxide (H(2)O(2)) is often considered synonymously with mitochondrial dysfunction. However, these oxidative species are signaling molecules in normal physiology so that a change in production or abundance is not a good criterion for mitochondrial dysfunction. In this review, we consider generation of reactive electrophiles and consequent modification of mitochondrial proteins as a means to define mitochondrial dysfunction. Accumulated evidence indicates that 4-hydroxynonenal (HNE) modification of proteins reflects mitochondrial dysfunction and provides an operational criterion for experimental definition of mitochondrial dysfunction. Improved means to detect and quantify mitochondrial HNE-protein adduct formation could allow its use for environmental healthrisk assessment. Furthermore, application of improved mass spectrometry-based proteomic methods will lead to further understanding of the critical targets contributing to disease risk.
Collapse
Affiliation(s)
- James R. Roede
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, Georgia
| | - Dean P. Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
99
|
Hung YC, Wang PW, Pan TL. Functional proteomics reveal the effect of Salvia miltiorrhiza aqueous extract against vascular atherosclerotic lesions. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2010; 1804:1310-21. [DOI: 10.1016/j.bbapap.2010.02.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2009] [Revised: 01/15/2010] [Accepted: 02/01/2010] [Indexed: 11/29/2022]
|
100
|
Arun P, Ariyannur PS, Moffett JR, Xing G, Hamilton K, Grunberg NE, Ives JA, Namboodiri AMA. Metabolic acetate therapy for the treatment of traumatic brain injury. J Neurotrauma 2010; 27:293-8. [PMID: 19803785 DOI: 10.1089/neu.2009.0994] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Patients suffering from traumatic brain injury (TBI) have decreased markers of energy metabolism, including N-acetylaspartate (NAA) and ATP. In the nervous system, NAA-derived acetate provides acetyl-CoA required for myelin lipid synthesis. Acetate can also be oxidized in mitochondria for the derivation of metabolic energy. In the current study, using the controlled cortical impact model of TBI in rats, we investigated the effects of the hydrophobic acetate precursor, glyceryltriacetate (GTA), as a method of delivering metabolizable acetate to the injured brain. We found that GTA administration significantly increased the levels of both NAA and ATP in the injured hemisphere 4 and 6 days after injury, and also resulted in significantly improved motor performance in rats 3 days after injury.
Collapse
Affiliation(s)
- Peethambaran Arun
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
| | | | | | | | | | | | | | | |
Collapse
|