1
|
Ritter A, Han J, Bianconi S, Henrich D, Marzi I, Leppik L, Weber B. The Ambivalent Role of miRNA-21 in Trauma and Acute Organ Injury. Int J Mol Sci 2024; 25:11282. [PMID: 39457065 PMCID: PMC11508407 DOI: 10.3390/ijms252011282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/14/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Since their initial recognition, miRNAs have been the subject of rising scientific interest. Especially in recent years, miRNAs have been recognized to play an important role in the mediation of various diseases, and further, their potential as biomarkers was recognized. Rising attention has also been given to miRNA-21, which has proven to play an ambivalent role as a biomarker. Responding to the demand for biomarkers in the trauma field, the present review summarizes the contrary roles of miRNA-21 in acute organ damage after trauma with a specific focus on the role of miRNA-21 in traumatic brain injury, spinal cord injury, cardiac damage, lung injury, and bone injury. This review is based on a PubMed literature search including the terms "miRNA-21" and "trauma", "miRNA-21" and "severe injury", and "miRNA-21" and "acute lung respiratory distress syndrome". The present summary makes it clear that miRNA-21 has both beneficial and detrimental effects in various acute organ injuries, which precludes its utility as a biomarker but makes it intriguing for mechanistic investigations in the trauma field.
Collapse
Affiliation(s)
- Aileen Ritter
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60486 Frankfurt am Main, Germany; (J.H.); (S.B.); (D.H.); (I.M.); (L.L.); (B.W.)
| | | | | | | | | | | | | |
Collapse
|
2
|
Boggs RC, Watts LT, Fox PT, Clarke GD. Metabolic Diaschisis in Mild Traumatic Brain Injury. J Neurotrauma 2024; 41:e1793-e1806. [PMID: 38482809 PMCID: PMC11564852 DOI: 10.1089/neu.2023.0290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024] Open
Abstract
Neurophysiological diaschisis presents in traumatic brain injury (TBI) as functional impairment distant to the lesion site caused by axonal neuroexcitation and deafferentation. Diaschisis studies in TBI models have evaluated acute phase functional and microstructural changes. Here, in vivo biochemical changes and cerebral blood flow (CBF) dynamics following TBI are studied with magnetic resonance. Behavioral assessments, magnetic resonance spectroscopy (MRS), and CBF measurements on rats followed cortical impact TBI. Data were acquired pre-TBI and 1-3 h, 2-days, 7-days, and 14-days post-TBI. MRS was performed on the ipsilateral and contralateral sides in the cortex, striatum, and thalamus. Metabolites measured by MRS included N-acetyl aspartate (NAA), aspartate (Asp), lactate (Lac), glutathione (GSH), and glutamate (Glu). Lesion volume expanded for 2 days post-TBI and then decreased. Ipsilateral CBF dropped acutely versus baseline on both sides (-62% ipsilateral, -48% contralateral, p < 0.05) but then recovered in cortex, with similar changes in ipsilateral striatum. Metabolic changes versus baseline included increased Asp (+640% by Day 7 post-TBI, p < 0.05) and Lac (+140% on Day 2 post-TBI, p < 0.05) in ipsilateral cortex, while GSH (-67% acutely, p < 0.05) and NAA decreased (-50% on Day 2, p < 0.05). In contralateral cortex Lac decreased (-73% acutely, p < 0.05). Analysis of variance showed significance for Side (p < 0.05), Time after TBI (p < 0.05), and interactions (p < 0.005) for Asp, GSH, Lac, and NAA. Transient decreases of GSH (-30%, p < 0.05, acutely) and NAA (-23% on Day 2, p < 0.05) occurred in ipsilateral striatum with reduced GSH (-42%, p < 0.005, acutely) in the contralateral striatum. GSH was decreased in ipsilateral thalamus (-59% ipsilateral on Day 2, p < 0.05). Delayed increases of total choline were seen in the contralateral thalamus were noted as well (+21% on Day 7 post-TBI, p < 0.05). Both CBF and neurometabolite concentration changes occurred remotely from the TBI site, both ipsilaterally and contralaterally. Decreased Lac levels on the contralateral cortex following TBI may be indicative of reduced anaerobic metabolism during the acute phase. The timing and locations of the changes suggest excitatory and inhibitory signaling processes are affecting post-TBI metabolic fluctuations.
Collapse
Affiliation(s)
- Robert C. Boggs
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Radiology and Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Lora T. Watts
- Department of Radiology and Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- Department of Anatomy, University of the Incarnate Word School of Osteopathic Medicine, San Antonio, Texas, USA
| | - Peter T. Fox
- Department of Radiology and Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Geoffrey D. Clarke
- Department of Radiology and Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
3
|
Huang YN, Greig NH, Huang PS, Chiang YH, Hoffer A, Yang CH, Tweedie D, Chen Y, Ou JC, Wang JY. Pomalidomide Improves Motor Behavioral Deficits and Protects Cerebral Cortex and Striatum Against Neurodegeneration Through a Reduction of Oxidative/Nitrosative Damages and Neuroinflammation After Traumatic Brain Injury. Cell Transplant 2024; 33:9636897241237049. [PMID: 38483119 PMCID: PMC10943757 DOI: 10.1177/09636897241237049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 12/06/2023] [Accepted: 12/12/2023] [Indexed: 03/18/2024] Open
Abstract
Neuronal damage resulting from traumatic brain injury (TBI) causes disruption of neuronal projections and neurotransmission that contribute to behavioral deficits. Cellular generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) is an early event following TBI. ROS often damage DNA, lipids, proteins, and carbohydrates while RNS attack proteins. The products of lipid peroxidation 4-hydroxynonenal (4-HNE) and protein nitration 3-nitrotyrosine (3-NT) are often used as indicators of oxidative and nitrosative damages, respectively. Increasing evidence has shown that striatum is vulnerable to damage from TBI with a disturbed dopamine neurotransmission. TBI results in neurodegeneration, oxidative stress, neuroinflammation, neuronal apoptosis, and autophagy in the striatum and contribute to motor or behavioral deficits. Pomalidomide (Pom) is a Food and Drug Administration (FDA)-approved immunomodulatory drug clinically used in treating multiple myeloma. We previously showed that Pom reduces neuroinflammation and neuronal death induced by TBI in rat cerebral cortex. Here, we further compared the effects of Pom in cortex and striatum focusing on neurodegeneration, oxidative and nitrosative damages, as well as neuroinflammation following TBI. Sprague-Dawley rats subjected to a controlled cortical impact were used as the animal model of TBI. Systemic administration of Pom (0.5 mg/kg, intravenous [i.v.]) at 5 h post-injury alleviated motor behavioral deficits, contusion volume at 24 h after TBI. Pom alleviated TBI-induced neurodegeneration stained by Fluoro-Jade C in both cortex and striatum. Notably, Pom treatment reduces oxidative and nitrosative damages in cortex and striatum and is more efficacious in striatum (93% reduction in 4-HNE-positive and 84% reduction in 3-NT-positive neurons) than in cerebral cortex (42% reduction in 4-HNE-positive and 55% reduction in 3-NT-positive neurons). In addition, Pom attenuated microgliosis, astrogliosis, and elevations of proinflammatory cytokines in cortical and striatal tissue. We conclude that Pom may contribute to improved motor behavioral outcomes after TBI through targeting oxidative/nitrosative damages and neuroinflammation.
Collapse
Affiliation(s)
- Ya-Ni Huang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei
- Department of Nursing, Hsin Sheng Junior College of Medical Care and Management, Taoyuan City
| | - Nigel H. Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Pen-Sen Huang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei
| | - Yung-Hsiao Chiang
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei
- Neuroscience Research Center, Taipei Medical University, Taipei
| | - Alan Hoffer
- Department of Neurosurgery, University Hospitals of Cleveland, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Chih-Hao Yang
- Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Ying Chen
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei
| | - Ju-Chi Ou
- Neuroscience Research Center, Taipei Medical University, Taipei
| | - Jia-Yi Wang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei
- Neuroscience Research Center, Taipei Medical University, Taipei
| |
Collapse
|
4
|
Schmitt R, Qayum S, Pliss A, Kuzmin AN, Muthaiah VPK, Kaliyappan K, Prasad PN, Mahajan SD. Mitochondrial Dysfunction and Apoptosis in Brain Microvascular Endothelial Cells Following Blast Traumatic Brain Injury. Cell Mol Neurobiol 2023; 43:3639-3651. [PMID: 37314617 DOI: 10.1007/s10571-023-01372-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/01/2023] [Indexed: 06/15/2023]
Abstract
Blood brain barrier (BBB) breakdown is a key driver of traumatic brain injury (TBI), contributing to prolonged neurological deficits and increased risk of death in TBI patients. Strikingly, the role of endothelium in the progression of BBB breakdown has not been sufficiently investigated, even though it constitutes the bulk of BBB structure. In the current study, we investigate TBI-induced changes in the brain endothelium at the subcellular level, particularly focusing on mitochondrial dysfunction, using a combination of confocal imaging, gene expression analysis, and molecular profiling by Raman spectrometry. Herein, we developed and applied an in-vitro blast-TBI (bTBI) model that employs an acoustic shock tube to deliver injury to cultured human brain microvascular endothelial cells (HBMVEC). We found that this injury results in aberrant expression of mitochondrial genes, as well as cytokines/ inflammasomes, and regulators of apoptosis. Furthermore, injured cells exhibit a significant increase in reactive oxygen species (ROS) and in Ca2+ levels. These changes are accompanied by overall reduction of intracellular proteins levels as well as profound transformations in mitochondrial proteome and lipidome. Finally, blast injury leads to a reduction in HBMVEC cell viability, with up to 50% of cells exhibiting signs of apoptosis following 24 h after injury. These findings led us to hypothesize that mitochondrial dysfunction in HBMVEC is a key component of BBB breakdown and TBI progression.
Collapse
Affiliation(s)
- Rebecca Schmitt
- Institute for Lasers, Photonics and Biophotonics, Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Sana Qayum
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, Clinical Translational Research Center, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Artem Pliss
- Institute for Lasers, Photonics and Biophotonics, Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Andrey N Kuzmin
- Institute for Lasers, Photonics and Biophotonics, Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Vijaya Prakash Krishnan Muthaiah
- Department of Rehabilitation Science, School of Public Health and Health Professions, The State University of New York, 633 Kimball Tower, Buffalo, NY, 14214, USA
| | - Kathiravan Kaliyappan
- Department of Rehabilitation Science, School of Public Health and Health Professions, The State University of New York, 633 Kimball Tower, Buffalo, NY, 14214, USA
| | - Paras N Prasad
- Institute for Lasers, Photonics and Biophotonics, Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA.
| | - Supriya D Mahajan
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, Clinical Translational Research Center, State University of New York at Buffalo, Buffalo, NY, 14203, USA.
| |
Collapse
|
5
|
Liu JJ, Long YF, Xu P, Guo HD, Cui GH. Pathogenesis of miR-155 on nonmodifiable and modifiable risk factors in Alzheimer's disease. Alzheimers Res Ther 2023; 15:122. [PMID: 37452431 PMCID: PMC10347850 DOI: 10.1186/s13195-023-01264-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 06/28/2023] [Indexed: 07/18/2023]
Abstract
Alzheimer's disease (AD) is a common age-related neurodegenerative disease in the central nervous system and is the primary cause of dementia. It is clinically characterized by the memory impairment, aphasia, apraxia, agnosia, visuospatial and executive dysfunction, behavioral changes, and so on. Incidence of this disease was bound up with age, genetic factors, cardiovascular and cerebrovascular dysfunction, and other basic diseases, but the exact etiology has not been clarified. MicroRNAs (miRNAs) are small endogenous non-coding RNAs that were involved in the regulation of post-transcriptional gene expression. miRNAs have been extensively studied as noninvasive potential biomarkers for disease due to their relative stability in bodily fluids. In addition, they play a significant role in the physiological and pathological processes of various neurological disorders, including stroke, AD, and Parkinson's disease. MiR-155, as an important pro-inflammatory mediator of neuroinflammation, was reported to participate in the progression of β-amyloid peptide and tau via regulating immunity and inflammation. In this review, we put emphasis on the effects of miR-155 on AD and explore the underlying biological mechanisms which could provide a novel approach for diagnosis and treatment of AD.
Collapse
Affiliation(s)
- Jia-Jia Liu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yun-Fan Long
- Department of Neurology, Shanghai No. 9 People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Peng Xu
- Affiliated Hospital of Jining Medical University, Jining, 272000, Shandong, China.
| | - Hai-Dong Guo
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Guo-Hong Cui
- Department of Neurology, Shanghai No. 9 People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
6
|
Yasmin A, Jokivarsi K, Poutiainen P, Pitkänen A, Gröhn O, Immonen R. Chronic hypometabolism in striatum and hippocampal network after traumatic brain injury and their relation with memory impairment - [18F]-FDG-PET and MRI 4 months after fluid percussion injury in rat. Brain Res 2022; 1788:147934. [PMID: 35483447 DOI: 10.1016/j.brainres.2022.147934] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 11/29/2022]
Abstract
Hippocampal and thalamo-cortico-striatal networks are critical for memory function as well as execution of a variety of learning strategies. In subjects with memory impairment as a sequel of traumatic brain injury (TBI), the contribution of late metabolic depression across these networks to memory deficit is poorly understood. We used [18F]-FDG-PET to measure chronic post-TBI glucose uptake in the striatum and connected brain areas (septal and temporal hippocampus, thalamus, entorhinal cortex, frontoparietal cortex and amygdala) in rats with lateral fluid-percussion injury (LFPI). Then we assessed a link between network hypometabolism and memory impairment. At 4 months post TBI, glucose uptake was decreased in ipsilateral striatum (10%, p = 0.027), frontoparietal cortex (17%, p = 0.00009), and hippocampus (22%, p = 0.027) as compared to sham operated controls. Thalamic uptake was 6% lower ipsilaterally than contralaterally, p = 0.00004). At 5 months, Morris water maze (MWM) showed memory impairment in 83% of the rats with TBI. The lower the hippocampal or striatal [18F]-FDG uptake, the poorer the MWM performance (hippocampus: r = -0.471, p < 0.05; striatum: r = -0.696, p < 0.001). Striatal [18F]-FDG-PET identified the injured animals with memory impairment with 100% specificity and sensitivity (AUC = 1.000, p = 0.009). Interestingly, the low striatal glucose uptake was a better diagnostic biomarker for memory impairment than the reduced hippocampal (AUC = 0.806, p = 0.112) or entorhinal (AUC = 0.528, p = 0.885) glucose uptake. The volumetric atrophy assessed in T2 weighted MRI or the gliotic area in Nissl staining did not correlate with glucose uptake. Arterial spin labeling did not indicate any reduction in the striatal blood flow. Our study suggests that TBI-induced chronic hypometabolism in striatum contributes to the cognitive deficits.
Collapse
Affiliation(s)
- Amna Yasmin
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Kimmo Jokivarsi
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Pekka Poutiainen
- Department of Radiopharmacy, Kuopio University Hospital, P.O. Box 100, FI-70029 KYS, Kuopio, Finland
| | - Asla Pitkänen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Olli Gröhn
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Riikka Immonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| |
Collapse
|
7
|
Zingale VD, Gugliandolo A, Mazzon E. MiR-155: An Important Regulator of Neuroinflammation. Int J Mol Sci 2021; 23:90. [PMID: 35008513 PMCID: PMC8745074 DOI: 10.3390/ijms23010090] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 12/18/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNA molecules that regulate gene expression at the post-transcriptional level and that play an important role in many cellular processes, including modulation of inflammation. MiRNAs are present in high concentrations in the central nervous system (CNS) and are spatially and temporally expressed in a specific way. Therefore, an imbalance in the expression pattern of these small molecules can be involved in the development of neurological diseases. Generally, CNS responds to damage or disease through the activation of an inflammatory response, but many neurological disorders are characterized by uncontrolled neuroinflammation. Many studies support the involvement of miRNAs in the activation or inhibition of inflammatory signaling and in the promotion of uncontrolled neuroinflammation with pathological consequences. MiR-155 is a pro-inflammatory mediator of the CNS and plays an important regulatory role. The purpose of this review is to summarize how miR-155 is regulated and the pathological consequences of its deregulation during neuroinflammatory disorders, including multiple sclerosis, Alzheimer's disease and other neuroinflammatory disorders. Modulation of miRNAs' expression could be used as a therapeutic strategy in the treatment of pathological neuroinflammation.
Collapse
Affiliation(s)
| | - Agnese Gugliandolo
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy; (V.D.Z.); (E.M.)
| | | |
Collapse
|
8
|
Balasubramanian N, Jadhav G, Sakharkar AJ. Repeated mild traumatic brain injuries perturb the mitochondrial biogenesis via DNA methylation in the hippocampus of rat. Mitochondrion 2021; 61:11-24. [PMID: 34508891 DOI: 10.1016/j.mito.2021.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 08/26/2021] [Accepted: 09/07/2021] [Indexed: 12/23/2022]
Abstract
Mitochondrial biogenesis in the brain is impaired in various neurological disorders including traumatic brain injury (TBI). The long-lasting effects of TBI may be, in part, attributed to epigenetic mechanisms such as DNA methylation. However, the role of DNA methylation on regulatory elements of nuclear and mitochondrial genome in mitochondrial biogenesis is not known. We examined the epigenetic regulation of mitochondrial transcription factor A (TFAM), and further probed its implications in mitochondrial dysfunction in the hippocampus of rats subjected to repeated mild TBI (rMTBI) using weight drop injury paradigm. rMTBI-induced hypermethylation at TFAM promoter resulted in deficits in its protein levels in mitochondria after immediate (48 h) and protracted (30 d) time points. Further, rMTBI also caused hypomethylation of mitochondrial DNA (mtDNA) promoters (HSP1 and HSP2), which further culminated into low binding of TFAM. rMTBI-induced changes weakened mitochondrial biogenesis in terms of reduced mtDNA-encoded rRNA, mRNA, and protein levels leading to shortages of ATP. To verify the potential role of mtDNA methylation in rMTBI-induced persistent mitochondrial dysfunction, rMTBI-induced rats were treated with methionine, a methyl donor. Methionine treatment restored the methylation levels on HSP1 and HSP2 resulting in efficient binding of TFAM and normalized the rRNA, mRNA, and protein levels. These findings suggest the crucial role of DNA methylation at nuclear and mitochondrial promoter regions in mitochondrial gene expression and ATP activity in the hippocampus after rMTBI.
Collapse
Affiliation(s)
| | - Gouri Jadhav
- Department of Biotechnology, Savitribai Phule Pune University, Pune 411 007, India
| | - Amul J Sakharkar
- Department of Biotechnology, Savitribai Phule Pune University, Pune 411 007, India.
| |
Collapse
|
9
|
miRNAs as Potential Biomarkers for Traumatic Brain Injury: Pathway From Diagnosis to Neurorehabilitation. J Head Trauma Rehabil 2020; 36:E155-E169. [PMID: 33201038 DOI: 10.1097/htr.0000000000000632] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Biomarkers that can advance precision neurorehabilitation of the traumatic brain injury (TBI) are needed. MicroRNAs (miRNAs) have biological properties that could make them well suited for playing key roles in differential diagnoses and prognoses and informing likelihood of responsiveness to specific treatments. OBJECTIVE To review the evidence of miRNA alterations after TBI and evaluate the state of science relative to potential neurorehabilitation applications of TBI-specific miRNAs. METHODS This scoping review includes 57 animal and human studies evaluating miRNAs after TBI. PubMed, Scopus, and Google Scholar search engines were used. RESULTS Gold standard analytic steps for miRNA biomarker assessment are presented. Published studies evaluating the evidence for miRNAs as potential biomarkers for TBI diagnosis, severity, natural recovery, and treatment-induced outcomes were reviewed including statistical evaluation. Growing evidence for specific miRNAs, including miR21, as TBI biomarkers is presented. CONCLUSIONS There is evidence of differential miRNA expression in TBI in both human and animal models; however, gaps need to be filled in terms of replication using rigorous, standardized methods to isolate a consistent set of miRNA changes. Longitudinal studies in TBI are needed to understand how miRNAs could be implemented as biomarkers in clinical practice.
Collapse
|
10
|
Davis CK, Vemuganti R. DNA damage and repair following traumatic brain injury. Neurobiol Dis 2020; 147:105143. [PMID: 33127471 DOI: 10.1016/j.nbd.2020.105143] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/09/2020] [Accepted: 10/23/2020] [Indexed: 01/05/2023] Open
Abstract
Traumatic brain injury (TBI) is known to promote significant DNA damage irrespective of age, sex, and species. Chemical as well as structural DNA modification start within minutes and persist for days after TBI. Although several DNA repair pathways are induced following TBI, the simultaneous downregulation of some of the genes and proteins of these pathways leads to an aberrant overall DNA repair process. In many instances, DNA damages escape even the most robust repair mechanisms, especially when the repair process becomes overwhelmed or becomes inefficient by severe or repeated injuries. The persisting DNA damage and/or lack of DNA repair contributes to long-term functional deficits. In this review, we discuss the mechanisms of TBI-induced DNA damage and repair. We further discussed the putative experimental therapies that target the members of the DNA repair process for improved outcome following TBI.
Collapse
Affiliation(s)
- Charles K Davis
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; William S. Middleton VA Hospital, Madison, WI, USA.
| |
Collapse
|
11
|
Thangavelu B, Wilfred BS, Johnson D, Gilsdorf JS, Shear DA, Boutté AM. Penetrating Ballistic-Like Brain Injury Leads to MicroRNA Dysregulation, BACE1 Upregulation, and Amyloid Precursor Protein Loss in Lesioned Rat Brain Tissues. Front Neurosci 2020; 14:915. [PMID: 33071724 PMCID: PMC7530327 DOI: 10.3389/fnins.2020.00915] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 08/07/2020] [Indexed: 12/22/2022] Open
Abstract
Severe traumatic brain injury (TBI) is a risk factor for neurodegenerative diseases. Yet, the molecular events involving dysregulated miRNAs that may be associated with protein degradation in the brain remains elusive. Quantitation of more than 800 miRNAs was conducted using rat ipsilateral coronal brain tissues collected 1, 3, or 7 days after penetrating ballistic-like brain injury (PBBI). As a control for each time-point, Sham-operated animals received craniotomy alone. Microarray and systems biology analysis indicated that the amplitude and complexity of miRNAs affected were greatest 7 day after PBBI. Arrays and Q-PCR inferred that dysregulation of miR-135a, miR-328, miR-29c, and miR-21 were associated with altered levels of beta-site amyloid precursor protein cleaving enzyme 1 (BACE1), PSEN1, PSEN2, and amyloid precursor protein (APP) genes. These events were followed by increased levels of mature BACE1 protein and concomitant loss of full length APP within 3–7 days, then elevation of amyloid beta (Aβ)-40 7 days after PBBI. This study indicates that miRNA arrays, coupled with systems biology, may be used to guide study design prior validation of miRNA dysregulation. Associative analysis of miRNAs, mRNAs, and proteins within a proposed pathway are poised for further validation as biomarkers and therapeutic targets relevant to TBI-induced APP loss and subsequent Aβ peptide generation during neurodegeneration.
Collapse
Affiliation(s)
- Bharani Thangavelu
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Bernard S Wilfred
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - David Johnson
- Department of Pathology and Area Laboratory Services, Landstuhl Regional Medical Center, Landstuhl, Germany
| | - Janice S Gilsdorf
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Deborah A Shear
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Angela M Boutté
- Brain Trauma Neuroprotection Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| |
Collapse
|
12
|
Balasubramanian N, Srivastava A, Pawar N, Sagarkar S, Sakharkar AJ. Repeated mild traumatic brain injury induces persistent variations in mitochondrial DNA copy number in mesocorticolimbic neurocircuitry of the rat. Neurosci Res 2020; 155:34-42. [DOI: 10.1016/j.neures.2019.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 12/18/2022]
|
13
|
Salman M, Tabassum H, Parvez S. Tannic Acid Provides Neuroprotective Effects Against Traumatic Brain Injury Through the PGC-1α/Nrf2/HO-1 Pathway. Mol Neurobiol 2020; 57:2870-2885. [PMID: 32399817 DOI: 10.1007/s12035-020-01924-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/22/2020] [Indexed: 12/30/2022]
Abstract
The present research was conducted to elucidate a possible molecular mechanism related to neuromodulatory effects of tannic acid (TA) supplementation against traumatic brain injury (TBI) in a rodent model. Oxidative damage and neuroinflammation play a critical role in TBI and lead to behavioral alterations and neuronal dysfunction and death. These changes suggest a potential avenue in neurotherapeutic intervention. The aim of the present study was to investigate the neuroprotective effects of TA and potential mechanism of these effects in a controlled cortical impact injury model of TBI in Wistar rats that were treated with TA (50 mg/kg body weight. i.p.) before 30 min and 6 and 18 h after TBI. TBI-induced rats were examined after 24 h for behavioral dysfunction, Nissl stain, lipid peroxidation rate, glutathione level, activities of antioxidant enzymes (catalase, glutathione S-transferase, glutathione peroxidase, and superoxide dismutase), the expression level of 4-hydroxynonenal, pro-inflammatory cytokines such as tumor necrosis factor alpha and interleukin-1 beta, as well as brain edema and immunoreactivity of glial fibrillary acidic protein. Results indicated that TA supplementation significantly modulated above mentioned alterations. Moreover, TA treatment effectively upregulated the protein expression of peroxisome proliferator-activated receptor gamma co-activator 1 alpha (PGC-1α) and nuclear factor-E2-related factor-2 (Nrf2) as well as mitochondrial transcription factor A and heme oxygenase-1 (HO-1) following TBI. Overall, our results suggest that TA effectively ameliorates the behavioral alterations, oxidative damage, mitochondrial impairment, and inflammation against TBI that may be attributed to activation of PGC-1α/Nrf-2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Mohd Salman
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Heena Tabassum
- Division of Biomedical Sciences, Indian Council of Medical Research, Ministry of Health and Family Welfare, Govt. of India, V. Ramalingaswamy Bhawan, P.O. Box No. 4911, New Delhi, 110029, India.
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
14
|
Song H, Chen M, Chen C, Cui J, Johnson CE, Cheng J, Wang X, Swerdlow RH, DePalma RG, Xia W, Gu Z. Proteomic Analysis and Biochemical Correlates of Mitochondrial Dysfunction after Low-Intensity Primary Blast Exposure. J Neurotrauma 2019; 36:1591-1605. [PMID: 30484371 PMCID: PMC6909772 DOI: 10.1089/neu.2018.6114] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Service members during military actions or combat training are frequently exposed to primary blasts by weaponry. Most studies have investigated moderate or severe brain injuries from blasts generating overpressures >100 kPa, whereas understanding the pathophysiology of low-intensity blast (LIB)-induced mild traumatic brain injury (mTBI) leading to neurological deficits remains elusive. Our recent studies, using an open-field LIB-induced mTBI mouse model with a peak overpressure at 46.6 kPa, demonstrated behavioral impairments and brain nanoscale damages, notably mitochondrial and axonal ultrastructural changes. In this study, we used tandem mass tagged (TMT) quantitative proteomics and bioinformatics analysis to seek insights into the molecular mechanisms underlying ultrastructural pathology. Changes in global- and phospho-proteomes were determined at 3 and 24 h and at 7 and 30 days post injury (DPI), in order to investigate the biochemical and molecular correlates of mitochondrial dysfunction. Results showed striking dynamic changes in a total of 2216 proteins and 459 phosphorylated proteins at vary time points after blast. Disruption of key canonical pathways included evidence of mitochondrial dysfunction, oxidative stress, axonal/cytoskeletal/synaptic dysregulation, and neurodegeneration. Bioinformatic analysis identified blast-induced trends in networks related to cellular growth/development/movement/assembly and cell-to-cell signaling interactions. With observations of proteomic changes, we found LIB-induced oxidative stress associated with mitochondrial dysfunction mainly at 7 and 30 DPI. These dysfunctions included impaired fission-fusion dynamics, diminished mitophagy, decreased oxidative phosphorylation, and compensated respiration-relevant enzyme activities. Insights on the early pathogenesis of primary LIB-induced brain damage provide a template for further characterization of its chronic effects, identification of potential biomarkers, and targets for intervention.
Collapse
Affiliation(s)
- Hailong Song
- Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri
| | - Mei Chen
- Bedford VA Medical Center, Bedford, Massachusetts
| | - Chen Chen
- Department of Computer Sciences, University of Missouri, Columbia, Missouri
| | - Jiankun Cui
- Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri
- Truman VA Hospital Research Service, Columbia, Missouri
| | - Catherine E. Johnson
- Department of Mining and Nuclear Engineering, Missouri University of Science and Technology, Rolla, Missouri
| | - Jianlin Cheng
- Department of Computer Sciences, University of Missouri, Columbia, Missouri
| | - Xiaowan Wang
- Department of Neurology, University of Kansas Alzheimer's Disease Center, University of Kansas Medical Center, Kansas City, Kansas
| | - Russell H. Swerdlow
- Department of Neurology, University of Kansas Alzheimer's Disease Center, University of Kansas Medical Center, Kansas City, Kansas
| | - Ralph G. DePalma
- Office of Research and Development, Department of Veterans Affairs, Washington, DC
- Norman Rich Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Weiming Xia
- Bedford VA Medical Center, Bedford, Massachusetts
| | - Zezong Gu
- Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri
- Truman VA Hospital Research Service, Columbia, Missouri
| |
Collapse
|
15
|
Henry RJ, Doran SJ, Barrett JP, Meadows VE, Sabirzhanov B, Stoica BA, Loane DJ, Faden AI. Inhibition of miR-155 Limits Neuroinflammation and Improves Functional Recovery After Experimental Traumatic Brain Injury in Mice. Neurotherapeutics 2019; 16:216-230. [PMID: 30225790 PMCID: PMC6361054 DOI: 10.1007/s13311-018-0665-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Micro-RNAs (miRs) are short, noncoding RNAs that negatively regulate gene expression at the post-transcriptional level and have been implicated in the pathophysiology of secondary damage after traumatic brain injury (TBI). Among miRs linked to inflammation, miR-155 has been implicated as a pro-inflammatory factor in a variety of organ systems. We examined the expression profile of miR-155, following experimental TBI (controlled cortical impact) in adult male C57Bl/6 mice, as well as the effects of acute or delayed administration of a miR-155 antagomir on post-traumatic neuroinflammatory responses and neurological recovery. Trauma robustly increased miR-155 expression in the injured cortex over 7 days. Similar TBI-induced miR-155 expression changes were also found in microglia/macrophages isolated from the injured cortex at 7 days post-injury. A miR-155 hairpin inhibitor (antagomir; 0.5 nmol), administered intracerebroventricularly (ICV) immediately after injury, attenuated neuroinflammatory markers at both 1 day and 7 days post-injury and reduced impairments in spatial working memory. Delayed ICV infusion of the miR-155 antagomir (0.5 nmol/day), beginning 24 h post-injury and continuing for 6 days, attenuated neuroinflammatory markers at 7 days post-injury and improved motor, but not cognitive, function through 28 days. The latter treatment limited NADPH oxidase 2 expression changes in microglia/macrophages in the injured cortex and reduced cortical lesion volume. In summary, TBI causes a robust and persistent neuroinflammatory response that is associated with increased miR-155 expression in microglia/macrophages, and miR-155 inhibition reduces post-traumatic neuroinflammatory responses and improves neurological recovery. Thus, miR-155 may be a therapeutic target for TBI-related neuroinflammation.
Collapse
Affiliation(s)
- Rebecca J. Henry
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD USA
| | - Sarah J. Doran
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD USA
| | - James P. Barrett
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD USA
| | - Victoria E. Meadows
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD USA
| | - Boris Sabirzhanov
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD USA
| | - Bogdan A. Stoica
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD USA
| | - David J. Loane
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD USA
- Department of Anesthesiology, University of Maryland School of Medicine, 655 West Baltimore Street, No. 6-011, Baltimore, MD 21201 USA
| | - Alan I. Faden
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD USA
- Department of Anesthesiology, University of Maryland School of Medicine, 685 West Baltimore Street, MSTF No. 6-02, Baltimore, MD 21201 USA
| |
Collapse
|
16
|
Carlson SW, Dixon CE. Lithium Improves Dopamine Neurotransmission and Increases Dopaminergic Protein Abundance in the Striatum after Traumatic Brain Injury. J Neurotrauma 2018; 35:2827-2836. [PMID: 29699444 PMCID: PMC6247981 DOI: 10.1089/neu.2017.5509] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Experimental models of traumatic brain injury (TBI) recapitulate secondary injury sequela and cognitive dysfunction reported in patients afflicted with a TBI. Impairments in neurotransmission are reported in multiple brain regions in the weeks following experimental TBI and may contribute to behavioral dysfunction. Formation of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex is an important mechanism for neurotransmitter exocytosis. We previously showed that lithium treatment attenuated hippocampal decreases in α-synuclein and VAMP2, enhanced SNARE complex formation, and improved cognitive performance after TBI. However, the effect of TBI on striatal SNARE complex formation is not known. We hypothesized lithium treatment would attenuate TBI-induced impairments in evoked dopamine release and increase the abundance of synaptic proteins associated with dopamine neurotransmission. The current study evaluated the effect of lithium (1 mmol/kg/day) administration on striatal evoked dopamine neurotransmission, SNARE complex formation, and proposed actions of lithium, including inhibition of GSK3β, assessment of synaptic marker protein abundance, and synaptic proteins important for dopamine synthesis and transport following controlled cortical impact (CCI). Sprague-Dawley rats were subjected to CCI or sham injury and treated daily with lithium chloride or vehicle for 7 days post-injury. We provide novel evidence that CCI reduces SNARE protein and SNARE complex abundance in the striatum at 1 week post-injury. Lithium administration improved evoked dopamine release and increased the abundance of α-synuclein, D2 receptor, and phosphorylated tyrosine hydroxylase in striatal synaptosomes post-injury. These findings show that lithium treatment attenuated dopamine neurotransmission deficits and increased the abundance of synaptic proteins important for dopamine signaling after TBI.
Collapse
Affiliation(s)
- Shaun W. Carlson
- Department of Neurological Surgery, Safar Center for Resuscitation Research, VA Pittsburgh Healthcare System, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - C. Edward Dixon
- Department of Neurological Surgery, Safar Center for Resuscitation Research, VA Pittsburgh Healthcare System, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
17
|
Neuroprotective effects of pifithrin-α against traumatic brain injury in the striatum through suppression of neuroinflammation, oxidative stress, autophagy, and apoptosis. Sci Rep 2018; 8:2368. [PMID: 29402897 PMCID: PMC5799311 DOI: 10.1038/s41598-018-19654-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 01/02/2018] [Indexed: 12/12/2022] Open
Abstract
Cortical and hippocampal neuronal damages caused by traumatic brain injury (TBI) are associated with motor and cognitive impairments; however, only little attention paid to the striatal damage. It is known that the p53 tumor-suppressor transcription factor participated in TBI-induced secondary brain damage. We investigated how the p53 inactivator pifithrin (PFT)-α affected TBI-induced striatal neuronal damage at 24 h post-injury. Sprague-Dawley rats subjected to a controlled cortical impact were used as TBI models. We observed that p53 mRNA significantly increased, whereas p53 protein expression was distributed predominantly in neurons but not in glia cells in striatum after TBI. PFT-α improved motor deficit following TBI. PFT-α suppressed TBI-induced striatal glial activation and expression of proinflammatory cytokines. PFT-α alleviated TBI-induced oxidative damage TBI induced autophagy was evidenced by increased protein expression of Beclin-1 and shift of microtubule-associated light chain (LC)3-I to LC3-II, and decreased p62. These effects were reduced by PFT-α. Post-injury PFT-α treatment reduced the number of degenerating (FJC-positive) and apoptotic neurons. Our results suggest that PFT-α may provide neuroprotective effects via p53-dependent or -independent mechanisms depending on the cell type and timing after the TBI and can possibly be developed into a novel therapy to ameliorate TBI-induced neuronal damage.
Collapse
|
18
|
Schiavone S, Neri M, Trabace L, Turillazzi E. The NADPH oxidase NOX2 mediates loss of parvalbumin interneurons in traumatic brain injury: human autoptic immunohistochemical evidence. Sci Rep 2017; 7:8752. [PMID: 28821783 PMCID: PMC5562735 DOI: 10.1038/s41598-017-09202-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/24/2017] [Indexed: 12/22/2022] Open
Abstract
Pharmacological interventions for traumatic brain injury (TBI) are limited. Together with parvalbumin (PV) loss, increased production of reactive oxygen species (ROS) by the NADPH oxidase NOX enzymes represents a key step in TBI. Here, we investigated the contribution of NOX2-derived oxidative stress to the loss of PV immunoreactivity associated to TBI, performing immunohistochemistry for NOX2, 8-hydroxy-2′-deoxyguanosine (8OHdG) and PV on post mortem brain samples of subjects died following TBI, subjects died from spontaneous intracerebral hemorrhage (SICH) and controls (CTRL). We detected an increased NOX2 expression and 8OHdG immunoreactivity in subjects died from TBI with respect to CTRL and SICH. NOX2 increase was mainly observed in GABAergic PV-positive interneurons, with a minor presence in microglia. No significant differences in other NADPH oxidase isoforms (NOX1 and NOX4) were detected among experimental groups. NOX2-derived oxidative stress elevation appeared a specific TBI-induced phenomenon, as no alterations in the nitrosative pathway were detected. Our results suggest that NOX2-derived oxidative stress might play a crucial role in the TBI-induced loss of PV-positive interneurons.
Collapse
Affiliation(s)
- Stefania Schiavone
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Margherita Neri
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy.
| | - Emanuela Turillazzi
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 20, 71122, Foggia, Italy
| |
Collapse
|
19
|
Gibbs WS, Weber RA, Schnellmann RG, Adkins DL. Disrupted mitochondrial genes and inflammation following stroke. Life Sci 2016; 166:139-148. [PMID: 27693381 DOI: 10.1016/j.lfs.2016.09.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 09/08/2016] [Accepted: 09/26/2016] [Indexed: 12/26/2022]
Abstract
AIMS Determine the subacute time course of mitochondria disruption, cell death, and inflammation in a rat model of unilateral motor cortical ischemic stroke. MAIN METHODS Rats received unilateral ischemia of the motor cortex and were tested on behavioral tasks to determine impairments. Animals were euthanized at 24h, 72h and 144h and mRNA expression of key mitochondria proteins and indicators of inflammation, apoptosis and potential regenerative processes in ipsilesion cortex and striatum, using RT-qPCR. Mitochondrial proteins were examined at 144h using immunoblot analysis. KEY FINDINGS Rats with stroke induced-behavioral deficits had sustained, 144h post-lesion, decreases in mitochondrial-encoded electron transport chain proteins NADH dehydrogenase subunit-1 and cytochrome c oxidase subunit-1 (mRNA and protein) and mitochondrial DNA content in perilesion motor and sensory cortex. Uncoupling-protein-2 gene expression, but not superoxide dismutase-2, remained elevated in ipsilateral cortex and striatum at this time. Cortical inflammatory cytokine, interleukin-6, was increased early and was followed by increased macrophage marker F4/80 after stroke. Cleaved caspase-3 activation was elevated in cortex and growth associated protein-43 was elevated in the cortex and striatum six days post-lesion. SIGNIFICANCE We identified a relationship between three disrupted pathways, (1) sustained loss of mitochondrial proteins and mitochondrial DNA copy number in the cortex linked to decreased mitochondrial gene transcription; (2) early inflammatory response mediated by interleukin- 6 followed by macrophages; (3) apoptosis in conjunction with the activation of regenerative pathways. The stroke-induced spatial and temporal profiles lay the foundation to target pharmacological therapeutics to these three pathways.
Collapse
Affiliation(s)
- Whitney S Gibbs
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston SC, United States
| | - Rachel A Weber
- Department of Neuroscience, Medical University of South Carolina, Charleston SC, United States
| | - Rick G Schnellmann
- Ralph H. Johnson VA Medical Center, Charleston, SC, United States; Department of Pharmacy & Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, United States.
| | - DeAnna L Adkins
- Department of Neuroscience, Medical University of South Carolina, Charleston SC, United States; Center of Biomedical Imaging, Medical University of South Carolina, Charleston SC, United States; Health Sciences and Research, College of Health Professions, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|