1
|
Murray KE, Ravula AR, Stiritz VA, Cominski TP, Delic V, Marín de Evsikova C, Rama Rao KV, Chandra N, Beck KD, Pfister BJ, Citron BA. Sex and Genotype Affect Mouse Hippocampal Gene Expression in Response to Blast-Induced Traumatic Brain Injury. Mol Neurobiol 2025:10.1007/s12035-025-04879-5. [PMID: 40178780 DOI: 10.1007/s12035-025-04879-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 03/21/2025] [Indexed: 04/05/2025]
Abstract
Blast-induced traumatic brain injury (bTBI) has been identified as an increasingly prevalent cause of morbidity and mortality in both military and civilian populations over the past few decades. Functional outcomes following bTBI vary widely among individuals, and chronic neurodegenerative effects including cognitive impairments can develop without effective diagnosis and treatment. Genetic predispositions and sex differences may affect gene expression changes in response to bTBI and influence an individual's probability of sustaining long-term damage or exhibiting resilience and tissue repair. Male and female mice from eight genetically diverse and distinct strains (129S1/SvImJ, A/J, C57BL/6J, CAST/EiJ, NOD/ShiLtJ, NZO/HlLtJ, PWK/PhJ, WSB/EiJ) which encompassed 90% of the genetic variability in commercially available laboratory mice were exposed to a single bTBI (180 kPa) using a well-established shock tube system. Subacute changes in hippocampal gene expression due to blast exposure were assessed using RNA-seq at 1-month post-injury. We identified patterns of dysregulation in gene ontology terms and canonical pathways related to mitochondrial function, ribosomal structure, synaptic plasticity, protein degradation, and intracellular signaling that varied by sex and/or strain, including significant changes in genes encoding respiratory complex I of the electron transport chain in male WSB/EiJ mice and the glutamatergic synapse across more than half of our groups. This study represents a multi-level examination of how genetic variability may influence response to bTBI and provides a foundation for the identification of potential therapeutic targets that could be modulated to improve the health of Veterans and others with histories of blast exposures.
Collapse
Affiliation(s)
- Kathleen E Murray
- Laboratory of Molecular Biology, Research & Development, U.S. Department of Veterans Affairs, VA New Jersey Health Care System, East Orange, NJ, USA
- School of Graduate Studies, Rutgers Health, Newark, NJ, USA
| | - Arun Reddy Ravula
- Molecular Neurotherapeutics Laboratory, Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Victoria A Stiritz
- Neurobehavioral Research Laboratory, Research & Development, U.S. Department of Veterans Affairs, VA New Jersey Health Care System, East Orange, NJ, USA
- School of Graduate Studies, Rutgers Health, Newark, NJ, USA
| | - Tara P Cominski
- Neurobehavioral Research Laboratory, Research & Development, U.S. Department of Veterans Affairs, VA New Jersey Health Care System, East Orange, NJ, USA
- Division of Life Sciences, School of Arts and Sciences, Rutgers University, New Brunswick, NJ, USA
| | - Vedad Delic
- Laboratory of Molecular Biology, Research & Development, U.S. Department of Veterans Affairs, VA New Jersey Health Care System, East Orange, NJ, USA
- School of Graduate Studies, Rutgers Health, Newark, NJ, USA
- Department of Pharmacology, Physiology & Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ, 07101, USA
| | - Caralina Marín de Evsikova
- Epigenetics and Functional Genomics Laboratory, Research & Development, U.S. Department of Veterans Affairs, Bay Pines VA Healthcare System, Bay Pines, FL, USA
- Department of Molecular Medicine, University of South Florida, Tampa, FL, USA
| | - Kakulavarapu V Rama Rao
- Center for Injury Biomechanics, Materials, and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Namas Chandra
- Center for Injury Biomechanics, Materials, and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Kevin D Beck
- Neurobehavioral Research Laboratory, Research & Development, U.S. Department of Veterans Affairs, VA New Jersey Health Care System, East Orange, NJ, USA
- School of Graduate Studies, Rutgers Health, Newark, NJ, USA
- Department of Pharmacology, Physiology & Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ, 07101, USA
| | - Bryan J Pfister
- Center for Injury Biomechanics, Materials, and Medicine, Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Bruce A Citron
- Laboratory of Molecular Biology, Research & Development, U.S. Department of Veterans Affairs, VA New Jersey Health Care System, East Orange, NJ, USA.
- School of Graduate Studies, Rutgers Health, Newark, NJ, USA.
- Department of Pharmacology, Physiology & Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ, 07101, USA.
| |
Collapse
|
2
|
Hosseini E, Sepehrinezhad A, Momeni J, Ascenzi BM, Gorji A, Sahab-Negah S. The Telencephalon. FROM ANATOMY TO FUNCTION OF THE CENTRAL NERVOUS SYSTEM 2025:401-427. [DOI: 10.1016/b978-0-12-822404-5.00014-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
3
|
Svirsky SE, Henchir J, Parry M, Holets E, Zhang T, Gittes GK, Carlson SW, Dixon CE. Viral-mediated increased hippocampal neurogranin modulate synapses at one month in a rat model of controlled cortical impact. Sci Rep 2024; 14:28998. [PMID: 39578516 PMCID: PMC11584851 DOI: 10.1038/s41598-024-77682-2] [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: 02/27/2024] [Accepted: 10/24/2024] [Indexed: 11/24/2024] Open
Abstract
Reductions of neurogranin (Ng), a calcium-sensitive calmodulin-binding protein, result in significant impairment across various hippocampal-dependent learning and memory tasks. Conversely, increasing levels of Ng facilitates synaptic plasticity, increases synaptogenesis and boosts cognitive abilities. Controlled cortical impact (CCI), an experimental traumatic brain injury (TBI) model, results in significantly reduced hippocampal Ng protein expression up to 4 weeks post-injury, supporting a strategy to increase Ng to improve function. In this study, hippocampal Ng expression was increased in adult, male Sham and CCI injured animals using intraparenchymal injection of adeno-associated virus (AAV) 30 min post-injury, thereby also affording the ability to differentiate endogenous and exogenous Ng. At 4 weeks, molecular, anatomical, and behavioral measures of synaptic plasticity were evaluated to determine the therapeutic potential of Ng modulation post-TBI. Increasing Ng had a TBI-dependent effect on hippocampal expression of synaptic proteins and dendritic spine morphology. Increasing Ng did not improve behavior across all outcomes in both Sham and CCI groups at the 4 week time-point. Overall, increasing Ng expression modulated protein expression and dendritic spine morphology, but exerted limited functional benefit after CCI. This study furthers our understanding of Ng, and mechanisms of cognitive dysfunction within the synapse sub-acutely after TBI.
Collapse
Affiliation(s)
- Sarah E Svirsky
- Center for Neuroscience, University of Pittsburgh, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Jeremy Henchir
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Madison Parry
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Erik Holets
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Ting Zhang
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - George K Gittes
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Shaun W Carlson
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - C Edward Dixon
- Center for Neuroscience, University of Pittsburgh, 4401 Penn Ave, Pittsburgh, PA, 15224, USA.
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA.
- V.A. Pittsburgh Healthcare System, 4401 Penn Ave, Pittsburgh, PA, 15224, USA.
| |
Collapse
|
4
|
Ghaderi S, Gholipour P, Safari S, Sadati SM, Brooshghalan SE, Sohrabi R, Rashidi K, Komaki A, Salehi I, Sarihi A, Zarei M, Shahidi S, Rashno M. Uncovering the protective potential of vanillic acid against traumatic brain injury-induced cognitive decline in male rats: Insights into underlying mechanisms. Biomed Pharmacother 2024; 179:117405. [PMID: 39236478 DOI: 10.1016/j.biopha.2024.117405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024] Open
Abstract
Traumatic brain injury (TBI) is a significant contributor to global mortality and disability, and there is still no specific drug available to treat cognitive deficits in survivors. Vanillic acid (VA), a bioactive phenolic compound, has shown protective effects in various models of neurodegeneration; however, its impact on TBI outcomes remains elusive. Therefore, this study aimed to elucidate the possible role of VA in ameliorating TBI-induced cognitive decline and to reveal the mechanisms involved. TBI was induced using the Marmarou impact acceleration model to deliver an impact force of 300 g, and treatment with VA (50 mg/kg; P.O.) was initiated 30 minutes post-TBI. The cognitive performance, hippocampal long-term potentiation (LTP), oxidative stress markers, neurological function, cerebral edema, and morphological changes were assessed at scheduled points in time. TBI resulted in cognitive decline in the passive avoidance task, impaired LTP in the perforant path-dentate gyrus (PP-DG) pathway, increased hippocampal oxidative stress, cerebral edema, neurological deficits, and neuronal loss in the rat hippocampus. In contrast, acute VA administration mitigated all the aforementioned TBI outcomes. The data suggest that reducing synaptic plasticity impairment, regulating oxidative and antioxidant defense, alleviating cerebral edema, and preventing neuronal loss by VA can be at least partially attributed to its protection against TBI-induced cognitive decline.
Collapse
Affiliation(s)
- Shahab Ghaderi
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Parsa Gholipour
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Samaneh Safari
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyed Mahdi Sadati
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Shahla Eyvari Brooshghalan
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rezvan Sohrabi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Khodabakhsh Rashidi
- Research Center of Oils and Fats, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Alireza Komaki
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Salehi
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Abdolrahman Sarihi
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Zarei
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Siamak Shahidi
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Masome Rashno
- Asadabad School of Medical Sciences, Asadabad, Iran.
| |
Collapse
|
5
|
Nikulina E, Tsokas P, Whitney K, Tcherepanov A, Hsieh C, Sacktor TC, Bergold PJ. Increased protein kinase Mζ expression by Minocycline and N-acetylcysteine restores late-phase long-term potentiation and spatial learning after closed head injury in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.20.613738. [PMID: 39345361 PMCID: PMC11429999 DOI: 10.1101/2024.09.20.613738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Cognitive deficits frequently arise after traumatic brain injury. The murine closed head injury (CHI) models these deficits since injured mice cannot acquire Barnes maze. Dosing of minocycline plus N-acetylcysteine beginning 12 hours post-CHI (MN12) restores Barnes maze acquisition by an unknown mechanism. Increased hippocampal synaptic efficacy is needed to acquire Barnes maze, synaptic long-term potentiation (LTP) models this increased synaptic efficacy in vitro . LTP has an early phase (E-LTP) lasting up to one hour that is mediated by second messengers that is followed by a late phase (L-LTP) that needs new synthesis of protein kinase M zeta (PKMζ). PKMζ has constitutive kinase activity because it lacks the autoinhibitory regulatory domain found in other PKCs. Due to its constitutive activity, the amount of PKMζ kinase activity is determined by PKMζ protein levels. We report that CHI bilaterally decreases PKMζ levels in the CA3 and CA1 hippocampus. MN12 increases CA1 PKMζ expression. CHI inhibits E-LTP in slices from the ipsilesional hippocampus and inhibits L-LTP in slices from both hippocamppi. MN12 treatment reestablishes both E-LTP and L-LTP in slices from the injured MN12-treated hippocampus. The restoration of L-LTP from injured MN12-treated hippocampus is mediated by PKMζ because L-LTP is blocked by the specific PKMζ inhibitor, ζ-stat. Hippocampal ζ-stat infusions also prevents Barnes maze acquisition in injured, MN12-treated mice. These data suggest that post-injury minocycline plus N-acetylcysteine targets PKMζ to improve synaptic plasticity and cognition in mice with closed-head injury.
Collapse
|
6
|
Svirsky SE, Henchir J, Li Y, Carlson SW, Dixon CE. Temporal-Specific Sex and Injury-Dependent Changes on Neurogranin-Associated Synaptic Signaling After Controlled Cortical Impact in Rats. Mol Neurobiol 2024; 61:7256-7268. [PMID: 38376763 DOI: 10.1007/s12035-024-04043-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 02/08/2024] [Indexed: 02/21/2024]
Abstract
Extensive effort has been made to study the role of synaptic deficits in cognitive impairment after traumatic brain injury (TBI). Neurogranin (Ng) is a calcium-sensitive calmodulin (CaM)-binding protein essential for Ca2+/CaM-dependent kinase II (CaMKII) autophosphorylation which subsequently modulates synaptic plasticity. Given the loss of Ng expression after injury, additional research is warranted to discern changes in hippocampal post-synaptic signaling after TBI. Under isoflurane anesthesia, adult, male and female Sprague-Dawley rats received a sham/control or controlled cortical impact (CCI) injury. Ipsilateral hippocampal synaptosomes were isolated at 24 h and 1, 2, and 4 weeks post-injury, and western blot was used to evaluate protein expression of Ng-associated signaling proteins. Non-parametric Mann-Whitney tests were used to determine significance of injury for each sex at each time point. There were significant changes in the hippocampal synaptic expression of Ng and associated synaptic proteins such as phosphorylated Ng, CaMKII, and CaM up to 4 weeks post-CCI, demonstrating TBI alters hippocampal post-synaptic signaling. This study furthers our understanding of mechanisms of cognitive dysfunction within the synapse sub-acutely after TBI.
Collapse
Affiliation(s)
- Sarah E Svirsky
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Jeremy Henchir
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Youming Li
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - Shaun W Carlson
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA
| | - C Edward Dixon
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Neurological Surgery, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA, 15224, USA.
- V.A. Pittsburgh Healthcare System, Pittsburgh, PA, USA.
| |
Collapse
|
7
|
Borucki DM, Rohrer B, Tomlinson S. Complement propagates visual system pathology following traumatic brain injury. J Neuroinflammation 2024; 21:98. [PMID: 38632569 PMCID: PMC11022420 DOI: 10.1186/s12974-024-03098-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is associated with the development of visual system disorders. Visual deficits can present with delay and worsen over time, and may be associated with an ongoing neuroinflammatory response that is known to occur after TBI. Complement system activation is strongly associated with the neuroinflammatory response after TBI, but whether it contributes to vision loss after TBI is unexplored. METHODS Acute and chronic neuroinflammatory changes within the dorsal lateral geniculate nucleus (dLGN) and retina were investigated subsequent to a moderate to severe murine unilateral controlled cortical impact. Neuroinflammatory and histopathological outcomes were interpreted in the context of behavioral and visual function data. To investigate the role of complement, cohorts were treated after TBI with the complement inhibitor, CR2-Crry. RESULTS At 3 days after TBI, complement component C3 was deposited on retinogeniculate synapses in the dLGN both ipsilateral and contralateral to the lesion, which was reduced in CR2-Crry treated animals. This was associated with microglia morphological changes in both the ipsilateral and contralateral dLGN, with a less ramified phenotype in vehicle compared to CR2-Crry treated animals. Microglia in vehicle treated animals also had a greater internalized VGlut2 + synaptic volume after TBI compared to CR2-Crry treated animals. Microglia morphological changes seen acutely persisted for at least 49 days after injury. Complement inhibition also reduced microglial synaptic internalization in the contralateral dLGN and increased the association between VGLUT2 and PSD95 puncta, indicating preservation of intact synapses. Unexpectedly, there were no changes in the thickness of the inner retina, retinal nerve fiber layer or retinal ganglion layer. Neuropathological changes in the dLGN were accompanied by reduced visual acuity at subacute and chronic time points after TBI, with improvement seen in CR2-Crry treated animals. CONCLUSION TBI induces complement activation within the dLGN and promotes microglial activation and synaptic internalization. Complement inhibition after TBI in a clinically relevant paradigm reduces complement activation, maintains a more surveillance-like microglia phenotype, and preserves synaptic density within the dLGN. Together, the data indicate that complement plays a key role in the development of visual deficits after TBI via complement-dependent microglial phagocytosis of synapses within the dLGN.
Collapse
Affiliation(s)
- Davis M Borucki
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Baerbel Rohrer
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC, USA.
- Ralph Johnson VA Medical Center, Charleston, SC, USA.
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.
- Ralph Johnson VA Medical Center, Charleston, SC, USA.
| |
Collapse
|
8
|
Fesharaki-Zadeh A, Datta D. An overview of preclinical models of traumatic brain injury (TBI): relevance to pathophysiological mechanisms. Front Cell Neurosci 2024; 18:1371213. [PMID: 38682091 PMCID: PMC11045909 DOI: 10.3389/fncel.2024.1371213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/20/2024] [Indexed: 05/01/2024] Open
Abstract
Background Traumatic brain injury (TBI) is a major cause of morbidity and mortality, affecting millions annually worldwide. Although the majority of TBI patients return to premorbid baseline, a subset of patient can develop persistent and often debilitating neurocognitive and behavioral changes. The etiology of TBI within the clinical setting is inherently heterogenous, ranging from sport related injuries, fall related injuries and motor vehicle accidents in the civilian setting, to blast injuries in the military setting. Objective Animal models of TBI, offer the distinct advantage of controlling for injury modality, duration and severity. Furthermore, preclinical models of TBI have provided the necessary temporal opportunity to study the chronic neuropathological sequelae of TBI, including neurodegenerative sequelae such as tauopathy and neuroinflammation within the finite experimental timeline. Despite the high prevalence of TBI, there are currently no disease modifying regimen for TBI, and the current clinical treatments remain largely symptom based. The preclinical models have provided the necessary biological substrate to examine the disease modifying effect of various pharmacological agents and have imperative translational value. Methods The current review will include a comprehensive survey of well-established preclinical models, including classic preclinical models including weight drop, blast injury, fluid percussion injury, controlled cortical impact injury, as well as more novel injury models including closed-head impact model of engineered rotational acceleration (CHIMERA) models and closed-head projectile concussive impact model (PCI). In addition to rodent preclinical models, the review will include an overview of other species including large animal models and Drosophila. Results There are major neuropathological perturbations post TBI captured in various preclinical models, which include neuroinflammation, calcium dysregulation, tauopathy, mitochondrial dysfunction and oxidative stress, axonopathy, as well as glymphatic system disruption. Conclusion The preclinical models of TBI continue to offer valuable translational insight, as well as essential neurobiological basis to examine specific disease modifying therapeutic regimen.
Collapse
Affiliation(s)
- Arman Fesharaki-Zadeh
- Department of Neurology and Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Dibyadeep Datta
- Division of Aging and Geriatric Psychiatry, Alzheimer’s Disease Research Unit, Department of Psychiatry, New Haven, CT, United States
| |
Collapse
|
9
|
Pordel S, McCloskey AP, Almahmeed W, Sahebkar A. The protective effects of statins in traumatic brain injury. Pharmacol Rep 2024; 76:235-250. [PMID: 38448729 DOI: 10.1007/s43440-024-00582-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024]
Abstract
Traumatic brain injury (TBI), often referred to as the "silent epidemic", is the most common cause of mortality and morbidity worldwide among all trauma-related injuries. It is associated with considerable personal, medical, and economic consequences. Although remarkable advances in therapeutic approaches have been made, current treatments and clinical management for TBI recovery still remain to be improved. One of the factors that may contribute to this gap is that existing therapies target only a single event or pathology. However, brain injury after TBI involves various pathological mechanisms, including inflammation, oxidative stress, blood-brain barrier (BBB) disruption, ionic disturbance, excitotoxicity, mitochondrial dysfunction, neuronal necrosis, and apoptosis. Statins have several beneficial pleiotropic effects (anti-excitotoxicity, anti-inflammatory, anti-oxidant, anti-thrombotic, immunomodulatory activity, endothelial and vasoactive properties) in addition to promoting angiogenesis, neurogenesis, and synaptogenesis in TBI. Supposedly, using agents such as statins that target numerous and diverse pathological mechanisms, may be more effective than a single-target approach in TBI management. The current review was undertaken to investigate and summarize the protective mechanisms of statins against TBI. The limitations of conducted studies and directions for future research on this potential therapeutic application of statins are also discussed.
Collapse
Affiliation(s)
- Safoora Pordel
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alice P McCloskey
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Wael Almahmeed
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
10
|
Borucki D, Rohrer B, Tomlinson S. Complement propagates visual system pathology following traumatic brain injury. RESEARCH SQUARE 2024:rs.3.rs-3970621. [PMID: 38464312 PMCID: PMC10925413 DOI: 10.21203/rs.3.rs-3970621/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Background Traumatic brain injury (TBI) is associated with the development of visual system disorders. Visual deficits can present with delay and worsen over time, and may be associated with an ongoing neuroinflammatory response that is known to occur after TBI. Complement activation is strongly associated with the neuroinflammatory response after TBI, but whether it contributes to vision loss after TBI is unexplored. Methods Acute and chronic neuroinflammatory changes within the dorsal lateral geniculate nucleus (dLGN) and retina were investigated subsequent to murine controlled unilateral cortical impact. Neuroinflammatory and histopathological data were interpreted in the context of behavioral and visual function data. To investigate the role of complement, cohorts were treated after TBI with the complement inhibitor, CR2-Crry. Results At 3 days after TBI, complement C3 was deposited on retinogeniculate synapses in the dLGN both ipsilateral and contralateral to the lesion, which was reduced in CR2-Crry treated animals. This was associated with microglia morphological changes in both the ipsilateral and contralateral dLGN, with a more amoeboid phenotype in vehicle compared to CR2-Crry treated animals. Microglia in vehicle treated animals also had a greater internalized VGlut2+ synaptic volume after TBI compared to CR2-Crry treated animals. Microglia morphological changes seen acutely persisted for at least 49 days after injury. Complement inhibition also reduced microglial synaptic internalization in the contralateral dLGN and increased the association between VGLUT2 and PSD95 puncta, indicating preservation of intact synapses. Unexpectedly, there were no changes in the thickness of the inner retina, retinal nerve fiber layer or retinal ganglion layer. Pathologies were accompanied by reduced visual acuity at subacute and chronic time points after TBI, with improvement seen in CR2-Crry treated animals. Conclusion TBI induces complement activation within the dLGN and promotes microglial activation and synaptic internalization. Complement inhibition after TBI in a clinically relevant paradigm reduces complement activation, maintains a more surveillance-like microglia phenotype, and preserves synaptic density within the dLGN. Together, the data indicate that complement plays a key role in the development of visual deficits after TBI via complement-dependent microglial phagocytosis of synapses within the dLGN.
Collapse
|
11
|
Chen CM, Gung PY, Ho YC, Hamdin CD, Yet SF. Probucol treatment after traumatic brain injury activates BDNF/TrkB pathway, promotes neuroregeneration and ameliorates functional deficits in mice. Br J Pharmacol 2023; 180:2605-2622. [PMID: 37263748 DOI: 10.1111/bph.16157] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 04/11/2023] [Accepted: 05/22/2023] [Indexed: 06/03/2023] Open
Abstract
BACKGROUND AND PURPOSE Traumatic brain injury (TBI) is a major cause of mortality and morbidity worldwide, yet pharmacotherapies for TBI are currently lacking. Neuroregeneration is important in brain repair and functional recovery. In this study, probucol, a cholesterol-lowering drug with established safety profiles, was examined for its therapeutic effects and neuroregenerative actions in TBI. EXPERIMENTAL APPROACH Male mice were subjected to the controlled cortical impact model of TBI, followed by daily administration of probucol. Neurological and cognitive functions were evaluated. Histological analyses of the neocortex and hippocampus were performed to detect the lesion, dendritic degeneration (microtubule-associated protein 2), synaptic density (synaptophysin), neurogenesis (doublecortin), brain-derived neurotrophic factor (BDNF) and tropomyosin receptor kinase B (TrkB) activation. Involvement of BDNF/TrkB pathway in probucol-mediated effects was examined in primary cultures of cortical neurons. KEY RESULTS Probucol reduced brain lesion volume, enhanced the recovery of body symmetry, improved motor function and attenuated memory dysfunction after TBI. Meanwhile, probucol promoted post-injury dendritic growth and synaptogenesis and increased hippocampal proliferating neuronal progenitor cells, along with the formation as well as the survival of newborn neurons. Moreover, probucol enhances BDNF expression and TrkB activation. In vitro, probucol promoted neurite outgrowth, which was inhibited by a selective TrkB antagonist ANA-12. CONCLUSIONS AND IMPLICATIONS Probucol enhanced functional restoration and ameliorated cognitive impairment after TBI by promoting post-injury neuronal remodelling and neurogenesis. Increased activation of BDNF/TrkB pathway by probucol, at least in part, contributed to the neuroregenerative effects of probucol. Together, it may be promising to repurpose probucol for TBI.
Collapse
Affiliation(s)
- Chen-Mei Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Pei-Yu Gung
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Yen-Chun Ho
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
- Cardiovascular Biology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Candra D Hamdin
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
- National Health Research Institutes & Department of Life Sciences, National Central University Joint Ph.D. Program in Biomedicine, Taoyuan City, Taiwan
| | - Shaw-Fang Yet
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
12
|
Kodali M, Madhu LN, Reger RL, Milutinovic B, Upadhya R, Attaluri S, Shuai B, Shankar G, Shetty AK. A single intranasal dose of human mesenchymal stem cell-derived extracellular vesicles after traumatic brain injury eases neurogenesis decline, synapse loss, and BDNF-ERK-CREB signaling. Front Mol Neurosci 2023; 16:1185883. [PMID: 37284464 PMCID: PMC10239975 DOI: 10.3389/fnmol.2023.1185883] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 04/28/2023] [Indexed: 06/08/2023] Open
Abstract
An optimal intranasal (IN) dose of human mesenchymal stem cell-derived extracellular vesicles (hMSC-EVs), 90 min post-traumatic brain injury (TBI), has been reported to prevent the evolution of acute neuroinflammation into chronic neuroinflammation resulting in the alleviation of long-term cognitive and mood impairments. Since hippocampal neurogenesis decline and synapse loss contribute to TBI-induced long-term cognitive and mood dysfunction, this study investigated whether hMSC-EV treatment after TBI can prevent hippocampal neurogenesis decline and synapse loss in the chronic phase of TBI. C57BL6 mice undergoing unilateral controlled cortical impact injury (CCI) received a single IN administration of different doses of EVs or the vehicle at 90 min post-TBI. Quantifying neurogenesis in the subgranular zone-granule cell layer (SGZ-GCL) through 5'-bromodeoxyuridine and neuron-specific nuclear antigen double labeling at ~2 months post-TBI revealed decreased neurogenesis in TBI mice receiving vehicle. However, in TBI mice receiving EVs (12.8 and 25.6 × 109 EVs), the extent of neurogenesis was matched to naive control levels. A similar trend of decreased neurogenesis was seen when doublecortin-positive newly generated neurons were quantified in the SGZ-GCL at ~3 months post-TBI. The above doses of EVs treatment after TBI also reduced the loss of pre-and post-synaptic marker proteins in the hippocampus and the somatosensory cortex. Moreover, at 48 h post-treatment, brain-derived neurotrophic factor (BDNF), phosphorylated extracellular signal-regulated kinase 1/2 (p-ERK1/2), and phosphorylated cyclic AMP response-element binding protein (p-CREB) levels were downregulated in TBI mice receiving the vehicle but were closer to naïve control levels in TBI mice receiving above doses of hMSC-EVs. Notably, improved BDNF concentration observed in TBI mice receiving hMSC-EVs in the acute phase was sustained in the chronic phase of TBI. Thus, a single IN dose of hMSC-EVs at 90 min post-TBI can ease TBI-induced declines in the BDNF-ERK-CREB signaling, hippocampal neurogenesis, and synapses.
Collapse
|
13
|
Wang Y, Wernersbach I, Strehle J, Li S, Appel D, Klein M, Ritter K, Hummel R, Tegeder I, Schäfer MKE. Early posttraumatic CSF1R inhibition via PLX3397 leads to time- and sex-dependent effects on inflammation and neuronal maintenance after traumatic brain injury in mice. Brain Behav Immun 2022; 106:49-66. [PMID: 35933030 DOI: 10.1016/j.bbi.2022.07.164] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/08/2022] [Accepted: 07/30/2022] [Indexed: 10/31/2022] Open
Abstract
BACKGROUND There is a need for early therapeutic interventions after traumatic brain injury (TBI) to prevent neurodegeneration. Microglia/macrophage (M/M) depletion and repopulation after treatment with colony stimulating factor 1 receptor (CSF1R) inhibitors reduces neurodegeneration. The present study investigates short- and long-term consequences after CSF1R inhibition during the early phase after TBI. METHODS Sex-matched mice were subjected to TBI and CSF1R inhibition by PLX3397 for 5 days and sacrificed at 5 or 30 days post injury (dpi). Neurological deficits were monitored and brain tissues were examined for histo- and molecular pathological markers. RNAseq was performed with 30 dpi TBI samples. RESULTS At 5 dpi, CSF1R inhibition attenuated the TBI-induced perilesional M/M increase and associated gene expressions by up to 50%. M/M attenuation did not affect structural brain damage at this time-point, impaired hematoma clearance, and had no effect on IL-1β expression. At 30 dpi, following drug discontinuation at 5 dpi and M/M repopulation, CSF1R inhibition attenuated brain tissue loss regardless of sex, as well as hippocampal atrophy and thalamic neuronal loss in male mice. Selected gene markers of brain inflammation and apoptosis were reduced in males but increased in females after early CSF1R inhibition as compared to corresponding TBI vehicle groups. Neurological outcome in behaving mice was almost not affected. RNAseq and gene set enrichment analysis (GSEA) of injured brains at 30 dpi revealed more genes associated with dendritic spines and synapse function after early CSF1R inhibition as compared to vehicle, suggesting improved neuronal maintenance and recovery. In TBI vehicle mice, GSEA showed high oxidative phosphorylation, oxidoreductase activity and ribosomal biogenesis suggesting oxidative stress and increased abundance of metabolically highly active cells. More genes associated with immune processes and phagocytosis in PLX3397 treated females vs males, suggesting sex-specific differences in response to early CSF1R inhibition after TBI. CONCLUSIONS M/M attenuation after CSF1R inhibition via PLX3397 during the early phase of TBI reduces long-term brain tissue loss, improves neuronal maintenance and fosters synapse recovery. Overall effects were not sex-specific but there is evidence that male mice benefit more than female mice.
Collapse
Affiliation(s)
- Yong Wang
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Isa Wernersbach
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Jenny Strehle
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Shuailong Li
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Dominik Appel
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Matthias Klein
- Institute for Immunology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Katharina Ritter
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Regina Hummel
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Goethe-University Frankfurt, Medical Faculty, Theodor Stern Kai 7, 60590 Frankfurt, Germany
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center, Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; Focus Program Translational Neurosciences (FTN) of the Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany; Research Center for Immunotherapy (FZI), Johannes Gutenberg-University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany.
| |
Collapse
|
14
|
Eyolfson E, Carr T, Fraunberger E, Khan A, Clark I, Mychasiuk R, Lohman AW. Repeated mild traumatic brain injuries in mice cause age- and sex-specific alterations in dendritic spine density. Exp Neurol 2022; 357:114172. [PMID: 35863503 DOI: 10.1016/j.expneurol.2022.114172] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/08/2022] [Accepted: 07/14/2022] [Indexed: 11/25/2022]
Abstract
Mild traumatic brain injuries (mTBI) plague the human population and their prevalence is increasing annually. More so, repeated mTBIs (RmTBI) are known to manifest and compound neurological deficits in vulnerable populations. Age at injury and sex are two important factors influencing RmTBI pathophysiology, but we continue to know little about the specific effects of RmTBI in youth and females. In this study, we directly quantified the effects of RmTBI on adolescent and adult, male and female mice, with a closed-head lateral impact model. We report age- and sex-specific neurobehavioural deficits in motor function and working memory, microglia responses to injury, and the subsequent changes in dendritic spine density in select brain regions. Specifically, RmTBI caused increased footslips in adult male mice as assessed in a beam walk assay and significantly reduced the time spent with a novel object in adolescent male and female mice. RmTBIs caused a significant reduction in microglia density in male mice in the motor cortex, but not female mice. Finally, RmTBI significantly reduced dendritic spine density in the agranular insular cortex (a region of the prefrontal cortex in mice) and increased dendritic spine density in the adolescent male motor cortex. Together, the data provided in this study sheds new light on the heterogeneity in RmTBI-induced behavioural, glial, and neuronal architecture changes dependent on age and sex.
Collapse
Affiliation(s)
- Eric Eyolfson
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada.
| | - Thomas Carr
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada.
| | - Erik Fraunberger
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada.
| | - Asher Khan
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada.
| | - Isabel Clark
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada.
| | - Richelle Mychasiuk
- Department of Psychology, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada; Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia.
| | - Alexander W Lohman
- Alberta Children's Hospital Research Institute (ACHRI), Calgary, AB, Canada; Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute (HBI), University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
15
|
Frankowski JC, Tierno A, Pavani S, Cao Q, Lyon DC, Hunt RF. Brain-wide reconstruction of inhibitory circuits after traumatic brain injury. Nat Commun 2022; 13:3417. [PMID: 35701434 PMCID: PMC9197933 DOI: 10.1038/s41467-022-31072-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 05/31/2022] [Indexed: 11/09/2022] Open
Abstract
Despite the fundamental importance of understanding the brain's wiring diagram, our knowledge of how neuronal connectivity is rewired by traumatic brain injury remains remarkably incomplete. Here we use cellular resolution whole-brain imaging to generate brain-wide maps of the input to inhibitory neurons in a mouse model of traumatic brain injury. We find that somatostatin interneurons are converted into hyperconnected hubs in multiple brain regions, with rich local network connections but diminished long-range inputs, even at areas not directly damaged. The loss of long-range input does not correlate with cell loss in distant brain regions. Interneurons transplanted into the injury site receive orthotopic local and long-range input, suggesting the machinery for establishing distant connections remains intact even after a severe injury. Our results uncover a potential strategy to sustain and optimize inhibition after traumatic brain injury that involves spatial reorganization of the direct inputs to inhibitory neurons across the brain.
Collapse
Affiliation(s)
- Jan C Frankowski
- Department of Anatomy & Neurobiology, University of California, Irvine, CA, 92697, USA
| | - Alexa Tierno
- Department of Anatomy & Neurobiology, University of California, Irvine, CA, 92697, USA.
| | - Shreya Pavani
- Department of Anatomy & Neurobiology, University of California, Irvine, CA, 92697, USA
| | - Quincy Cao
- Department of Anatomy & Neurobiology, University of California, Irvine, CA, 92697, USA
| | - David C Lyon
- Department of Anatomy & Neurobiology, University of California, Irvine, CA, 92697, USA
| | - Robert F Hunt
- Department of Anatomy & Neurobiology, University of California, Irvine, CA, 92697, USA. .,Epilepsy Research Center, University of California, Irvine, CA, 92697, USA. .,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, 92697, USA. .,Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, 92697, USA. .,Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
16
|
Larson K, Damon M, Randhi R, Nixon-Lee N, J Dixon K. Selective inhibition of soluble TNF using XPro1595 improves hippocampal pathology to promote improved neurological recovery following traumatic brain injury in mice. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 22:CNSNDDT-EPUB-124336. [PMID: 35692164 DOI: 10.2174/1871527321666220610104908] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 06/15/2023]
Abstract
AIMS To determine the efficacy of XPro1595 to improve pathophysiological and functional outcomes in a mouse model of traumatic brain injury (TBI). BACKGROUND Symptoms associated with TBI can be debilitating, and treatment without off-target side effects remains a challenge. This study aimed to investigate the efficacy of selectively inhibiting the soluble form of TNF (solTNF) using the biologic XPro1595 in a mouse model of TBI. OBJECTIVES Use XPro1595 to determine whether injury-induced solTNF promotes hippocampal inflammation and dendritic plasticity, and associated functional impairments. METHODS Mild-to-moderate traumatic brain injury (CCI model) was induced in adult male C57Bl/6J WT and Thy1-YFPH mice, with XPro1595 (10 mg/kg, S.C.) or vehicle being administered in a clinically relevant window (60 minutes post-injury). The animals were assessed for differences in neurological function, and hippocampal tissue was analyzed for inflammation and glial reactivity, as well as neuronal degeneration and plasticity. RESULTS We report that unilateral CCI over the right parietal cortex in mice promoted deficits in learning and memory, depressive-like behavior, and neuropathic pain. Using immunohistochemical and Western blotting techniques, we observed the cortical injury promoted a set of expected pathophysiology's within the hippocampus consistent with the observed neurological outcomes, including glial reactivity, enhanced neuronal dendritic degeneration (dendritic beading), and reduced synaptic plasticity (spine density and PSD-95 expression) within the DG and CA1 region of the hippocampus, that were prevented in mice treated with XPro1595. CONCLUSION Overall, we observed that selectively inhibiting solTNF using XPro1595 improved the pathophysiological and neurological sequelae of brain-injured mice, which provides support for its use in patients with TBI.
Collapse
Affiliation(s)
- Katelyn Larson
- Department of Surgery, Virginia Commonwealth University, United States
| | - Melissa Damon
- Department of Surgery, Virginia Commonwealth University, United States
| | - Rajasa Randhi
- Department of Surgery, Virginia Commonwealth University, United States
| | - Nancy Nixon-Lee
- Department of Surgery, Virginia Commonwealth University, United States
| | - Kirsty J Dixon
- Department of Surgery, Virginia Commonwealth University, United States
| |
Collapse
|
17
|
Urrutia-Ruiz C, Rombach D, Cursano S, Gerlach-Arbeiter S, Schoen M, Bockmann J, Demestre M, Boeckers TM. Deletion of the Autism-Associated Protein SHANK3 Abolishes Structural Synaptic Plasticity after Brain Trauma. Int J Mol Sci 2022; 23:ijms23116081. [PMID: 35682760 PMCID: PMC9181590 DOI: 10.3390/ijms23116081] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 12/14/2022] Open
Abstract
Autism spectrum disorders (ASDs) are characterized by repetitive behaviors and impairments of sociability and communication. About 1% of ASD cases are caused by mutations of SHANK3, a major scaffolding protein of the postsynaptic density. We studied the role of SHANK3 in plastic changes of excitatory synapses within the central nervous system by employing mild traumatic brain injury (mTBI) in WT and Shank3 knockout mice. In WT mice, mTBI triggered ipsi- and contralateral loss of hippocampal dendritic spines and excitatory synapses with a partial recovery over time. In contrast, no significant synaptic alterations were detected in Shank3∆11−/− mice, which showed fewer dendritic spines and excitatory synapses at baseline. In line, mTBI induced the upregulation of synaptic plasticity-related proteins Arc and p-cofilin only in WT mice. Interestingly, microglia proliferation was observed in WT mice after mTBI but not in Shank3∆11−/− mice. Finally, we detected TBI-induced increased fear memory at the behavioral level, whereas in Shank3∆11−/− animals, the already-enhanced fear memory levels increased only slightly after mTBI. Our data show the lack of structural synaptic plasticity in Shank3 knockout mice that might explain at least in part the rigidity of behaviors, problems in adjusting to new situations and cognitive deficits seen in ASDs.
Collapse
Affiliation(s)
- Carolina Urrutia-Ruiz
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
| | - Daniel Rombach
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
| | - Silvia Cursano
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
| | - Susanne Gerlach-Arbeiter
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
| | - Michael Schoen
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
| | - Juergen Bockmann
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
| | - Maria Demestre
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
| | - Tobias M. Boeckers
- Institute for Anatomy and Cell Biology, Albert Einstein Allee 11, 89081 Ulm, Germany; (C.U.-R.); (D.R.); (S.C.); (S.G.-A.); (M.S.); (J.B.); (M.D.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Translational Biochemistry, 89081 Ulm, Germany
- Correspondence: ; Tel.: +49-731-5002-3220
| |
Collapse
|
18
|
Xu Y, Liu Z, Xu S, Li C, Li M, Cao S, Sun Y, Dai H, Guo Y, Chen X, Liang W. Scientific Evidences of Calorie Restriction and Intermittent Fasting for Neuroprotection in Traumatic Brain Injury Animal Models: A Review of the Literature. Nutrients 2022; 14:1431. [PMID: 35406044 PMCID: PMC9002547 DOI: 10.3390/nu14071431] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 12/11/2022] Open
Abstract
It has widely been accepted that food restriction (FR) without malnutrition has multiple health benefits. Various calorie restriction (CR) and intermittent fasting (IF) regimens have recently been reported to exert neuroprotective effects in traumatic brain injury (TBI) through variable mechanisms. However, the evidence connecting CR or IF to neuroprotection in TBI as well as current issues remaining in this research field have yet to be reviewed in literature. The objective of our review was therefore to weigh the evidence that suggests the connection between CR/IF with recovery promotion following TBI. Medline, Google Scholar and Web of Science were searched from inception to 25 February 2022. An overwhelming number of results generated suggest that several types of CR/IF play a promising role in promoting post-TBI recovery. This recovery is believed to be achieved by alleviating mitochondrial dysfunction, promoting hippocampal neurogenesis, inhibiting glial cell responses, shaping neural cell plasticity, as well as targeting apoptosis and autophagy. Further, we represent our views on the current issues and provide thoughts on the future direction of this research field.
Collapse
Affiliation(s)
- Yang Xu
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China; (Y.X.); (S.X.); (C.L.); (Y.S.)
| | - Zejie Liu
- Department of Forensic Pathology and Forensic Clinical Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China; (Z.L.); (H.D.)
| | - Shuting Xu
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China; (Y.X.); (S.X.); (C.L.); (Y.S.)
| | - Chengxian Li
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China; (Y.X.); (S.X.); (C.L.); (Y.S.)
| | - Manrui Li
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China; (M.L.); (S.C.)
| | - Shuqiang Cao
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China; (M.L.); (S.C.)
| | - Yuwen Sun
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China; (Y.X.); (S.X.); (C.L.); (Y.S.)
| | - Hao Dai
- Department of Forensic Pathology and Forensic Clinical Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China; (Z.L.); (H.D.)
| | - Yadong Guo
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha 410013, China;
| | - Xiameng Chen
- Department of Forensic Pathology and Forensic Clinical Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China; (Z.L.); (H.D.)
| | - Weibo Liang
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China; (M.L.); (S.C.)
| |
Collapse
|
19
|
Williams HC, Carlson SW, Saatman KE. A role for insulin-like growth factor-1 in hippocampal plasticity following traumatic brain injury. VITAMINS AND HORMONES 2022; 118:423-455. [PMID: 35180936 DOI: 10.1016/bs.vh.2021.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Traumatic brain injury (TBI) initiates a constellation of secondary injury cascades, leading to neuronal damage and dysfunction that is often beyond the scope of endogenous repair mechanisms. Cognitive deficits are among the most persistent morbidities resulting from TBI, necessitating a greater understanding of mechanisms of posttraumatic hippocampal damage and neuroplasticity and identification of therapies that improve recovery by enhancing repair pathways. Focusing here on hippocampal neuropathology associated with contusion-type TBIs, the impact of brain trauma on synaptic structure and function and the process of adult neurogenesis is discussed, reviewing initial patterns of damage as well as evidence for spontaneous recovery. A case is made that insulin-like growth factor-1 (IGF-1), a growth-promoting peptide synthesized in both the brain and the periphery, is well suited to augment neuroplasticity in the injured brain. Essential during brain development, multiple lines of evidence delineate roles in the adult brain for IGF-1 in the maintenance of synapses, regulation of neurotransmission, and modulation of forms of synaptic plasticity such as long-term potentiation. Further, IGF-1 enhances adult hippocampal neurogenesis though effects on proliferation and neuronal differentiation of neural progenitor cells and on dendritic growth of newly born neurons. Post-injury administration of IGF-1 has been effective in rodent models of TBI in improving learning and memory, attenuating death of mature hippocampal neurons and promoting neurogenesis, providing critical proof-of-concept data. More studies are needed to explore the effects of IGF-1-based therapies on synaptogenesis and synaptic plasticity following TBI and to optimize strategies in order to stimulate only appropriate, functional neuroplasticity.
Collapse
Affiliation(s)
- Hannah C Williams
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, Lexington, KY, United States
| | - Shaun W Carlson
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kathryn E Saatman
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, Lexington, KY, United States.
| |
Collapse
|
20
|
Frankowski JC, Foik AT, Tierno A, Machhor JR, Lyon DC, Hunt RF. Traumatic brain injury to primary visual cortex produces long-lasting circuit dysfunction. Commun Biol 2021; 4:1297. [PMID: 34789835 PMCID: PMC8599505 DOI: 10.1038/s42003-021-02808-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 10/25/2021] [Indexed: 01/20/2023] Open
Abstract
Primary sensory areas of the mammalian neocortex have a remarkable degree of plasticity, allowing neural circuits to adapt to dynamic environments. However, little is known about the effects of traumatic brain injury on visual circuit function. Here we used anatomy and in vivo electrophysiological recordings in adult mice to quantify neuron responses to visual stimuli two weeks and three months after mild controlled cortical impact injury to primary visual cortex (V1). We found that, although V1 remained largely intact in brain-injured mice, there was ~35% reduction in the number of neurons that affected inhibitory cells more broadly than excitatory neurons. V1 neurons showed dramatically reduced activity, impaired responses to visual stimuli and weaker size selectivity and orientation tuning in vivo. Our results show a single, mild contusion injury produces profound and long-lasting impairments in the way V1 neurons encode visual input. These findings provide initial insight into cortical circuit dysfunction following central visual system neurotrauma.
Collapse
Affiliation(s)
- Jan C. Frankowski
- grid.266093.80000 0001 0668 7243Department of Anatomy & Neurobiology, University of California, Irvine, CA 92697 USA
| | - Andrzej T. Foik
- grid.413454.30000 0001 1958 0162Ophthalmic Biology Group, International Centre for Translational Eye Research, Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Alexa Tierno
- grid.266093.80000 0001 0668 7243Department of Anatomy & Neurobiology, University of California, Irvine, CA 92697 USA
| | - Jiana R. Machhor
- grid.266093.80000 0001 0668 7243Department of Anatomy & Neurobiology, University of California, Irvine, CA 92697 USA
| | - David C. Lyon
- grid.266093.80000 0001 0668 7243Department of Anatomy & Neurobiology, University of California, Irvine, CA 92697 USA
| | - Robert F. Hunt
- grid.266093.80000 0001 0668 7243Department of Anatomy & Neurobiology, University of California, Irvine, CA 92697 USA
| |
Collapse
|
21
|
Faillot M, Chaillet A, Palfi S, Senova S. Rodent models used in preclinical studies of deep brain stimulation to rescue memory deficits. Neurosci Biobehav Rev 2021; 130:410-432. [PMID: 34437937 DOI: 10.1016/j.neubiorev.2021.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 11/28/2022]
Abstract
Deep brain stimulation paradigms might be used to treat memory disorders in patients with stroke or traumatic brain injury. However, proof of concept studies in animal models are needed before clinical translation. We propose here a comprehensive review of rodent models for Traumatic Brain Injury and Stroke. We systematically review the histological, behavioral and electrophysiological features of each model and identify those that are the most relevant for translational research.
Collapse
Affiliation(s)
- Matthieu Faillot
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France
| | - Antoine Chaillet
- Laboratoire des Signaux et Systèmes (L2S-UMR8506) - CentraleSupélec, Université Paris Saclay, Institut Universitaire de France, France
| | - Stéphane Palfi
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France
| | - Suhan Senova
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France.
| |
Collapse
|
22
|
Parker KN, Donovan MH, Smith K, Noble-Haeusslein LJ. Traumatic Injury to the Developing Brain: Emerging Relationship to Early Life Stress. Front Neurol 2021; 12:708800. [PMID: 34484104 PMCID: PMC8416304 DOI: 10.3389/fneur.2021.708800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/22/2021] [Indexed: 12/01/2022] Open
Abstract
Despite the high incidence of brain injuries in children, we have yet to fully understand the unique vulnerability of a young brain to an injury and key determinants of long-term recovery. Here we consider how early life stress may influence recovery after an early age brain injury. Studies of early life stress alone reveal persistent structural and functional impairments at adulthood. We consider the interacting pathologies imposed by early life stress and subsequent brain injuries during early brain development as well as at adulthood. This review outlines how early life stress primes the immune cells of the brain and periphery to elicit a heightened response to injury. While the focus of this review is on early age traumatic brain injuries, there is also a consideration of preclinical models of neonatal hypoxia and stroke, as each further speaks to the vulnerability of the brain and reinforces those characteristics that are common across each of these injuries. Lastly, we identify a common mechanistic trend; namely, early life stress worsens outcomes independent of its temporal proximity to a brain injury.
Collapse
Affiliation(s)
- Kaila N. Parker
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| | - Michael H. Donovan
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| | - Kylee Smith
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| | - Linda J. Noble-Haeusslein
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
23
|
Whitney K, Nikulina E, Rahman SN, Alexis A, Bergold PJ. Delayed dosing of minocycline plus N-acetylcysteine reduces neurodegeneration in distal brain regions and restores spatial memory after experimental traumatic brain injury. Exp Neurol 2021; 345:113816. [PMID: 34310944 DOI: 10.1016/j.expneurol.2021.113816] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 07/07/2021] [Accepted: 07/20/2021] [Indexed: 11/16/2022]
Abstract
Multiple drugs to treat traumatic brain injury (TBI) have failed clinical trials. Most drugs lose efficacy as the time interval increases between injury and treatment onset. Insufficient therapeutic time window is a major reason underlying failure in clinical trials. Few drugs have been developed with therapeutic time windows sufficiently long enough to treat TBI because little is known about which brain functions can be targeted if therapy is delayed hours to days after injury. We identified multiple injury parameters that are improved by first initiating treatment with the drug combination minocycline (MINO) plus N-acetylcysteine (NAC) at 72 h after injury (MN72) in a mouse closed head injury (CHI) experimental TBI model. CHI produces spatial memory deficits resulting in impaired performance on Barnes maze, hippocampal neuronal loss, and bilateral damage to hippocampal neurons, dendrites, spines and synapses. MN72 treatment restores Barnes maze acquisition and retention, protects against hippocampal neuronal loss, limits damage to dendrites, spines and synapses, and accelerates recovery of microtubule associated protein 2 (MAP2) expression, a key protein in maintaining proper dendritic architecture and synapse density. These data show that in addition to the structural integrity of the dendritic arbor, spine and synapse density can be successfully targeted with drugs first dosed days after injury. Retention of substantial drug efficacy even when first dosed 72 h after injury makes MINO plus NAC a promising candidate to treat clinical TBI.
Collapse
Affiliation(s)
- Kristen Whitney
- Department of Physiology and Pharmacology, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America; Program in Neural and Behavioral Science, School of Graduate Studies, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America
| | - Elena Nikulina
- Department of Physiology and Pharmacology, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America
| | - Syed N Rahman
- Department of Physiology and Pharmacology, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America
| | - Alisia Alexis
- Department of Physiology and Pharmacology, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America
| | - Peter J Bergold
- Department of Physiology and Pharmacology, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America; Program in Neural and Behavioral Science, School of Graduate Studies, State University of New York-Downstate Health Sciences University, Brooklyn, NY 11215, United States of America.
| |
Collapse
|
24
|
Zhang Q, Liu F, Yan W, Wu Y, Wang M, Wei J, Wang S, Zhu X, Chai X, Zhao S. Prolonged maternal separation alters neurogenesis and synaptogenesis in postnatal dentate gyrus of mice. Bipolar Disord 2021; 23:376-390. [PMID: 32805776 DOI: 10.1111/bdi.12986] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVES As a common model for adverse early experience and depression, maternal separation (MS) is always used to investigate the psychological disease. Despite extensive and strong evidence verified the depression-like state induced by MS, little is known about the specific mechanism of MS. Therefore, the present study aimed to investigate the neurobiology mechanism of the MS-induced depression-like state. METHODS To verify the depression-like behaviors of offspring induced by MS, a series of behavioral tests were performed. Then, in vivo electroporation and three-dimensional reconstruction, combining with immunohistochemistry and BrdU labeling, were mainly used to explore the neurogenesis and synaptogenesis in postnatal dentate gyrus. RESULTS Prolonged MS indeed induced the depression-like behaviors of offspring in adulthood. Surprisingly, learning and memory were enhanced by prolonged MS. Further investigation indicated that prolonged MS inhibited the proliferation of neural stem cells, impaired the survival, and altered the fate decision of newborn cells, whereas the total length and terminal tips of dendrite, and the spine density, especially thin spine, were significantly increased in prolonged MS mice. CONCLUSIONS Our results elucidated that prolonged MS induced the depression-like state by impairing postnatal neurogenesis of dentate gyrus. Importantly, our results emphasized that prolonged MS increased the spine density, especially thin spine, by increasing the total length and number of terminal tips of dendrite, thereby enhancing learning and memory.
Collapse
Affiliation(s)
- Qianru Zhang
- College of Veterinary Medicine, Department of Neurobiology, Northwest A&F University, Yangling, China
| | - Feng Liu
- College of Veterinary Medicine, Department of Neurobiology, Northwest A&F University, Yangling, China
| | - Wenyong Yan
- College of Veterinary Medicine, Department of Neurobiology, Northwest A&F University, Yangling, China
| | - Yongji Wu
- College of Veterinary Medicine, Department of Neurobiology, Northwest A&F University, Yangling, China
| | - Mengli Wang
- College of Veterinary Medicine, Department of Neurobiology, Northwest A&F University, Yangling, China
| | - Jingjing Wei
- College of Veterinary Medicine, Department of Neurobiology, Northwest A&F University, Yangling, China
| | - Shuzhong Wang
- College of Veterinary Medicine, Department of Neurobiology, Northwest A&F University, Yangling, China
| | - Xiaoyan Zhu
- College of Veterinary Medicine, Department of Neurobiology, Northwest A&F University, Yangling, China
| | - Xuejun Chai
- College of Basic Medicine, Xi'an Medical University, Xi'an, China
| | - Shanting Zhao
- College of Veterinary Medicine, Department of Neurobiology, Northwest A&F University, Yangling, China
| |
Collapse
|
25
|
Qiu X, Ping S, Kyle M, Chin L, Zhao LR. SCF + G-CSF treatment in the chronic phase of severe TBI enhances axonal sprouting in the spinal cord and synaptic pruning in the hippocampus. Acta Neuropathol Commun 2021; 9:63. [PMID: 33832542 PMCID: PMC8028149 DOI: 10.1186/s40478-021-01160-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 03/17/2021] [Indexed: 12/19/2022] Open
Abstract
Traumatic brain injury (TBI) is a major cause of long-term disability in young adults. An evidence-based treatment for TBI recovery, especially in the chronic phase, is not yet available. Using a severe TBI mouse model, we demonstrate that the neurorestorative efficacy of repeated treatments with stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) (SCF + G-CSF) in the chronic phase is superior to SCF + G-CSF single treatment. SCF + G-CSF treatment initiated at 3 months post-TBI enhances contralesional corticospinal tract sprouting into the denervated side of the cervical spinal cord and re-balances the TBI-induced overgrown synapses in the hippocampus by enhancing microglial function of synaptic pruning. These neurorestorative changes are associated with SCF + G-CSF-improved somatosensory-motor function and spatial learning. In the chronic phase of TBI, severe TBI-caused microglial degeneration in the cortex and hippocampus is ameliorated by SCF + G-CSF treatment. These findings reveal the therapeutic potential and possible mechanism of SCF + G-CSF treatment in brain repair during the chronic phase of severe TBI.
Collapse
|
26
|
Jamjoom AAB, Rhodes J, Andrews PJD, Grant SGN. The synapse in traumatic brain injury. Brain 2021; 144:18-31. [PMID: 33186462 PMCID: PMC7880663 DOI: 10.1093/brain/awaa321] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability worldwide and is a risk factor for dementia later in life. Research into the pathophysiology of TBI has focused on the impact of injury on the neuron. However, recent advances have shown that TBI has a major impact on synapse structure and function through a combination of the immediate mechanical insult and the ensuing secondary injury processes, leading to synapse loss. In this review, we highlight the role of the synapse in TBI pathophysiology with a focus on the confluence of multiple secondary injury processes including excitotoxicity, inflammation and oxidative stress. The primary insult triggers a cascade of events in each of these secondary processes and we discuss the complex interplay that occurs at the synapse. We also examine how the synapse is impacted by traumatic axonal injury and the role it may play in the spread of tau after TBI. We propose that astrocytes play a crucial role by mediating both synapse loss and recovery. Finally, we highlight recent developments in the field including synapse molecular imaging, fluid biomarkers and therapeutics. In particular, we discuss advances in our understanding of synapse diversity and suggest that the new technology of synaptome mapping may prove useful in identifying synapses that are vulnerable or resistant to TBI.
Collapse
Affiliation(s)
- Aimun A B Jamjoom
- Centre for Clinical Brain Sciences, Chancellor's Building, Edinburgh BioQuarter, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Jonathan Rhodes
- Anaesthesia, Critical Care and Pain Medicine, University of Edinburgh, Edinburgh EH16 4SA, UK
| | - Peter J D Andrews
- Anaesthesia, Critical Care and Pain Medicine, University of Edinburgh, Edinburgh EH16 4SA, UK
| | - Seth G N Grant
- Centre for Clinical Brain Sciences, Chancellor's Building, Edinburgh BioQuarter, University of Edinburgh, Edinburgh EH16 4SB, UK
- Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| |
Collapse
|
27
|
Carlson SW, Yan HQ, Li Y, Henchir J, Ma X, Young MS, Ikonomovic MD, Dixon CE. Differential Regional Responses in Soluble Monomeric Alpha Synuclein Abundance Following Traumatic Brain Injury. Mol Neurobiol 2021; 58:362-374. [PMID: 32948930 PMCID: PMC7704579 DOI: 10.1007/s12035-020-02123-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 09/05/2020] [Indexed: 12/14/2022]
Abstract
Alpha synuclein (α-synuclein) is a neuronal protein found predominately in presynaptic terminals. While the pathological effect of α-synuclein aggregates has been a topic of intense study in several neurodegenerative conditions, less attention has been placed on changes in monomeric α-synuclein and related physiological consequences on neuronal function. A growing body of evidence supports an important physiological role of α-synuclein in neurotransmission. In the context of traumatic brain injury (TBI), we hypothesized that the regional abundance of soluble monomeric α-synuclein is altered over a chronic time period post-injury. To this end, we evaluated α-synuclein in the cortex, hippocampus, and striatum of adult rats at 6 h, 1 day, 1, 2, 4, and 8 weeks after controlled cortical impact (CCI) injury. Western blot analysis demonstrated decreased levels of monomer α-synuclein protein in the ipsilateral hippocampus at 6 h, 1 day, 1, 2, and 8 weeks, as well as in the ipsilateral cortex at 1 and 2 weeks and in the ipsilateral striatum at 6 h after CCI compared with sham animals. Immunohistochemical analysis revealed lower α-synuclein and a modest reduction in synaptophysin staining in the ipsilateral hippocampus at 1 week after CCI compared with sham animals, with no evidence of intracellular or extracellular α-synuclein aggregates. Collectively, these findings demonstrate that monomeric α-synuclein protein abundance in the hippocampus is reduced over an extensive (acute-to-chronic) post-injury interval. This deficit may contribute to the chronically impaired neurotransmission known to occur after TBI.
Collapse
Affiliation(s)
- S W Carlson
- Neurological Surgery, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - H Q Yan
- Neurological Surgery, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - Y Li
- Neurological Surgery, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - J Henchir
- Neurological Surgery, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - X Ma
- Neurological Surgery, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - M S Young
- Neurological Surgery, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| | - M D Ikonomovic
- Neurology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA, 15261, USA
- VA Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - C E Dixon
- Neurological Surgery, University of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.
- VA Pittsburgh Healthcare System, Pittsburgh, PA, USA.
| |
Collapse
|
28
|
Segovia-Oropeza M, Santiago-Castañeda C, Orozco-Suárez SA, Concha L, Rocha L. Sodium Cromoglycate Decreases Sensorimotor Impairment and Hippocampal Alterations Induced by Severe Traumatic Brain Injury in Rats. J Neurotrauma 2020; 37:2595-2603. [PMID: 32484040 DOI: 10.1089/neu.2019.6975] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Severe traumatic brain injury (TBI) results in significant functional disturbances in the hippocampus. Studies support that sodium cromoglycate (CG) induces neuroprotective effects. This study focused on investigating the effects of post-TBI subchronic administration of CG on hippocampal hyperexcitability and damage as well as on sensorimotor impairment in rats. In contrast to the control group (Sham+SS group), animals undergoing severe TBI (TBI+SS group) showed sensorimotor dysfunction over the experimental post-TBI period (day 2, 55%, p < 0.001; day 23, 39.5%, p < 0.001; day 30, 38.6%, p < 0.01). On day 30 post-TBI, TBI+SS group showed neuronal hyperexcitability (63.3%, p < 0.01). The hippocampus ipsilateral to the injury showed volume reduction (14.4%, p < 0.001) with a volume of damage of 0.15 ± 0.09 mm3. These changes were associated with neuronal loss in the dentate gyrus (ipsilateral, 33%, p < 0.05); hilus (ipsilateral, 77%, p < 0.001; contralateral, 51%, p < 0.001); Cornu Ammonis (CA)1 (ipsilateral, 40%, p < 0.01), and CA3 (ipsilateral, 52%, p < 0.001; contralateral, 34%, p < 0.01). Animals receiving subchronic treatment with CG (50 mg/kg, s.c. daily for 10 days) after TBI (TBI+CG group) displayed a sensorimotor dysfunction less evident than that of the TBI+SS group (p < 0.001). Their hippocampal excitability was similar to that of the Sham+SS group (p = 0.21). The TBI+CG group presented hippocampal volume reduction (12.7%, p = 0.94) and damage (0.10 ± 0.03 mm3, p > 0.99) similar to the TBI+SS group. However, their hippocampal neuronal preservation was similar to that of the Sham+SS group. These results indicate that CG represents an appropriate and novel pharmacological strategy to reduce the long-term sensorimotor impairment and hippocampal damage and hyperexcitability that result as consequences of severe TBI.
Collapse
Affiliation(s)
| | | | | | - Luis Concha
- Institute of Neurobiology, National Autonomous University of Mexico, Campus Juriquilla, Queretaro, Mexico
| | - Luisa Rocha
- Department of Pharmacobiology, Center of Research and Advanced Studies, Mexico City, Mexico
| |
Collapse
|
29
|
An update on the association between traumatic brain injury and Alzheimer's disease: Focus on Tau pathology and synaptic dysfunction. Neurosci Biobehav Rev 2020; 120:372-386. [PMID: 33171143 DOI: 10.1016/j.neubiorev.2020.10.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/09/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023]
Abstract
L.P. Li, J.W. Liang and H.J. Fu. An update on the association between traumatic brain injury and Alzheimer's disease: Focus on Tau pathology and synaptic dysfunction. NEUROSCI BIOBEHAV REVXXX-XXX,2020.-Traumatic brain injury (TBI) and Alzheimer's disease (AD) are devastating conditions that have long-term consequences on individual's cognitive functions. Although TBI has been considered a risk factor for the development of AD, the link between TBI and AD is still in debate. Aggregation of hyperphosphorylated tau and intercorrelated synaptic dysfunction, two key pathological elements in both TBI and AD, play a pivotal role in mediating neurodegeneration and cognitive deficits, providing a mechanistic link between these two diseases. In the first part of this review, we analyze the experimental literatures on tau pathology in various TBI models and review the distribution, biological features and mechanisms of tau pathology following TBI with implications in AD pathogenesis. In the second part, we review evidences of TBI-mediated structural and functional impairments in synapses, with a focus on the overlapped mechanisms underlying synaptic abnormalities in both TBI and AD. Finally, future perspectives are proposed for uncovering the complex relationship between TBI and neurodegeneration, and developing potential therapeutic avenues for alleviating cognitive deficits after TBI.
Collapse
|
30
|
Traumatic Brain Injury Preserves Firing Rates But Disrupts Laminar Oscillatory Coupling and Neuronal Entrainment in Hippocampal CA1. eNeuro 2020; 7:ENEURO.0495-19.2020. [PMID: 32737188 PMCID: PMC7477953 DOI: 10.1523/eneuro.0495-19.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 07/15/2020] [Accepted: 07/19/2020] [Indexed: 11/21/2022] Open
Abstract
While hippocampal-dependent learning and memory are particularly vulnerable to traumatic brain injury (TBI), the functional status of individual hippocampal neurons and their interactions with oscillations are unknown following injury. Using the most common rodent TBI model and laminar recordings in CA1, we found a significant reduction in oscillatory input into the radiatum layer of CA1 after TBI. Surprisingly, CA1 neurons maintained normal firing rates despite attenuated input, but did not maintain appropriate synchronization with this oscillatory input or with local high-frequency oscillations. Normal synchronization between these coordinating oscillations was also impaired. Simultaneous recordings of medial septal neurons known to participate in theta oscillations revealed increased GABAergic/glutamatergic firing rates postinjury under anesthesia, potentially because of a loss of modulating feedback from the hippocampus. These results suggest that TBI leads to a profound disruption of connectivity and oscillatory interactions, potentially disrupting the timing of CA1 neuronal ensembles that underlie aspects of learning and memory.
Collapse
|
31
|
Sauerbeck AD, Gangolli M, Reitz SJ, Salyards MH, Kim SH, Hemingway C, Gratuze M, Makkapati T, Kerschensteiner M, Holtzman DM, Brody DL, Kummer TT. SEQUIN Multiscale Imaging of Mammalian Central Synapses Reveals Loss of Synaptic Connectivity Resulting from Diffuse Traumatic Brain Injury. Neuron 2020; 107:257-273.e5. [PMID: 32392471 PMCID: PMC7381374 DOI: 10.1016/j.neuron.2020.04.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 03/04/2020] [Accepted: 04/11/2020] [Indexed: 02/07/2023]
Abstract
The brain's complex microconnectivity underlies its computational abilities and vulnerability to injury and disease. It has been challenging to illuminate the features of this synaptic network due to the small size and dense packing of its elements. Here, we describe a rapid, accessible super-resolution imaging and analysis workflow-SEQUIN-that quantifies central synapses in human tissue and animal models, characterizes their nanostructural and molecular features, and enables volumetric imaging of mesoscale synaptic networks without the production of large histological arrays. Using SEQUIN, we identify cortical synapse loss resulting from diffuse traumatic brain injury, a highly prevalent connectional disorder. Similar synapse loss is observed in three murine models of Alzheimer-related neurodegeneration, where SEQUIN mesoscale mapping identifies regional synaptic vulnerability. These results establish an easily implemented and robust nano-to-mesoscale synapse quantification and characterization method. They furthermore identify a shared mechanism-synaptopathy-between Alzheimer neurodegeneration and its best-established epigenetic risk factor, brain trauma.
Collapse
Affiliation(s)
- Andrew D Sauerbeck
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mihika Gangolli
- McKelvey School of Engineering, Washington University, St. Louis, MO 63130, USA; Currently, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Sydney J Reitz
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Maverick H Salyards
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Samuel H Kim
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christopher Hemingway
- Institute of Clinical Neuroimmunology, Ludwig-Maximilians Universität München, Munich 82152, Germany
| | - Maud Gratuze
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tejaswi Makkapati
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, Ludwig-Maximilians Universität München, Munich 82152, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich 81377, Germany
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David L Brody
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Currently, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Terrance T Kummer
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
32
|
Ratliff WA, Delic V, Pick CG, Citron BA. Dendritic arbor complexity and spine density changes after repetitive mild traumatic brain injury and neuroprotective treatments. Brain Res 2020; 1746:147019. [PMID: 32681835 DOI: 10.1016/j.brainres.2020.147019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/25/2020] [Accepted: 07/10/2020] [Indexed: 12/17/2022]
Abstract
Traumatic brain injury has been described as the signature affliction of recent military conflicts and repetitive TBIs, particularly associated with military and athletic activities, typically result in more severe clinical effects. The majority of TBIs are mild, but they can result in long term cognitive deficits for which there is no effective treatment. One of the most significant deficits observed in TBI patients is memory loss, which suggests that TBI can induce pathological changes within the hippocampus. tert-butylhydroquinone (tBHQ) and pioglitazone activate the Nrf2 and PPAR-γ transcription factors, respectively, and both have been shown to be neuroprotective in model systems. We examined the morphological changes within the hippocampus following repetitive mild TBI and simultaneous treatment with both factors. We utilized a closed head injury mouse model with five injuries over 5 weeks. Our results showed marked morphological changes among the dendrites and dendritic spines of the neurons of the dentate gyrus of the hippocampus. We observed decreases in overall dendritic length, as well as in the quantity and density of dendritic spines. Our treatment partially ameliorated these effects, suggesting that the Nrf2 and PPAR-γ transcription factors may be important targets for future drug development in the treatment of TBI in humans.
Collapse
Affiliation(s)
- Whitney A Ratliff
- Laboratory of Molecular Biology, Bay Pines VA Healthcare System, Research and Development 151, Bldg. 22 Rm. 123, 10000 Bay Pines Blvd, Bay Pines, FL 33744, United States
| | - Vedad Delic
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research & Development (15), Bldg. 16, Rm 16-176 385 Tremont Ave, East Orange, NJ 07018, United States
| | - Chaim G Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel; Dr. Miriam and Sheldon G. Adelson Chair and Center for the Biology of Addictive Diseases, Tel Aviv University, Tel Aviv, Israel
| | - Bruce A Citron
- Laboratory of Molecular Biology, VA New Jersey Health Care System, Research & Development (15), Bldg. 16, Rm 16-176 385 Tremont Ave, East Orange, NJ 07018, United States; Laboratory of Molecular Biology, Bay Pines VA Healthcare System, Research and Development 151, Bldg. 22 Rm. 123, 10000 Bay Pines Blvd, Bay Pines, FL 33744, United States; Department of Pharmacology, Physiology & Neuroscience, Rutgers-New Jersey Medical School, Newark, NJ 07103, United States.
| |
Collapse
|
33
|
Özevren H, Deveci E, Tuncer MC. The effect of rosmarinic acid on deformities occurring in brain tissue by craniectomy method. Histopathological evaluation of IBA-1 and GFAP expressions. Acta Cir Bras 2020; 35:e202000406. [PMID: 32578724 PMCID: PMC7307720 DOI: 10.1590/s0102-865020200040000006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/09/2020] [Accepted: 03/04/2020] [Indexed: 12/20/2022] Open
Abstract
PURPOSE To investigate the role of Rosmarinic acid (RA) in the prevention of traumatic brain injury and the immunohistochemical analysis of IBA-1 and GFAP expressions. METHODS Healthy male rats were randomly divided into 3 groups consisting of 10 rats. Groups were as follows; control group, traumatic brain injury (TBI) group, and TBI+RA group. After traumatic brain injury, blood samples were taken from the animals and analyzed with various biochemical markers. And then IBA-1 and GFAP expressions were evaluated immunohistochemically. RESULTS Significant results were obtained in all biochemical parameters between groups. Immunohistochemical sections showed IBA-1 not only in microglia and macrophage activity but also in degenerative neurons in blood vessel endothelial cells. However, GFAP reaction and post-traumatic rosmarinic acid administration showed positive expression in astrocytes with regular structure around the blood vessel. CONCLUSION Rosmarinic acid in blood vessel endothelial cells showed that preserving the integrity of astrocytic structure in the blood brain barrier may be an important antioxidant.
Collapse
Affiliation(s)
- Hüseyin Özevren
- Associate Professor, Department of Neurosurgery , Faculty of
Medicine , Dicle University , Diyarbakır , Turkey . Technical procedures, manuscript
preparation and writing, final approval
| | - Engin Deveci
- PhD, Professor, Department of Histology and Embryology , Faculty
of Medicine , Dicle University , Diyarbakır , Turkey . Technical procedures,
histopathological examinations, manuscript preparation and writing, final
approval
| | - Mehmet Cudi Tuncer
- PhD, Professor, Department of Anatomy , Faculty of Medicine ,
Dicle University , Diyarbakır , Turkey . Technical procedures, histopathological
examinations, manuscript preparation and writing, final approval
| |
Collapse
|
34
|
Qiu X, Ping S, Kyle M, Chin L, Zhao LR. Long-term beneficial effects of hematopoietic growth factors on brain repair in the chronic phase of severe traumatic brain injury. Exp Neurol 2020; 330:113335. [PMID: 32360282 DOI: 10.1016/j.expneurol.2020.113335] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 04/17/2020] [Accepted: 04/27/2020] [Indexed: 11/19/2022]
Abstract
Severe traumatic brain injury (TBI) is the major cause of long-term, even life-long disability and cognitive impairments in young adults. The lack of therapeutic approaches to improve recovery in the chronic phase of severe TBI is a big challenge to the medical research field. Using a single severe TBI model in young adult mice, this study examined the restorative efficacy of two hematopoietic growth factors, stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF), on brain repair in the chronic phase of TBI. SCF and G-CSF alone or combination (SCF + G-CSF) treatment was administered at 3 months post-TBI. Functional recovery was evaluated by neurobehavioral tests during the period of 21 weeks after treatment. Neuropathology was examined 22 weeks after treatment. We observed that severe TBI caused persistent impairments in spatial learning/memory and somatosensory-motor function, long-term and widespread neuropathology, including dendritic reduction, decrease and overgrowth of axons, over-generated excitatory synapses, and demyelination in the cortex, hippocampus and striatum. SCF, G-CSF, and SCF + G-CSF treatments ameliorated severe TBI-induced widespread neuropathology. SCF + G-CSF treatment showed superior efficacy in improving long-term functional outcome, enhancing neural plasticity, rebalancing neural structure networks disturbed by severe TBI, and promoting remyelination. These novel findings demonstrate the therapeutic potential of SCF and G-CSF in enhancing recovery in the chronic phase of severe TBI .
Collapse
Affiliation(s)
- Xuecheng Qiu
- Department of Neurosurgery, The State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Suning Ping
- Department of Neurosurgery, The State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Michele Kyle
- Department of Neurosurgery, The State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Lawrence Chin
- Department of Neurosurgery, The State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Li-Ru Zhao
- Department of Neurosurgery, The State University of New York Upstate Medical University, Syracuse, NY 13210, USA; VA Health Care Upstate New York, Syracuse VA Medical Center, USA.
| |
Collapse
|
35
|
Svirsky S, Henchir J, Li Y, Ma X, Carlson S, Dixon CE. Neurogranin Protein Expression Is Reduced after Controlled Cortical Impact in Rats. J Neurotrauma 2020; 37:939-949. [PMID: 31691647 PMCID: PMC7175627 DOI: 10.1089/neu.2019.6759] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Traumatic brain injury (TBI) is known to cause short- and long-term synaptic changes in the brain, possibly underlying downstream cognitive impairments. Neuronal levels of neurogranin, a calcium-sensitive calmodulin-binding protein essential for synaptic plasticity and postsynaptic signaling, are correlated with cognitive function. This study aims to understand the effect of TBI on neurogranin by characterizing changes in protein expression at various time points after injury. Adult, male rats were subjected to either controlled cortical impact (CCI) or control surgery. Expression of neurogranin and post-synaptic density 95 (PSD-95) were evaluated by Western blot in the cortex and hippocampus at 24 h and 1, 2, and 4 weeks post-injury. We hypothesized that CCI reduces neurogranin levels in the cortex and hippocampus, and demonstrate different expression patterns from PSD-95. Neurogranin levels were reduced in the ipsilateral cortex and hippocampus up to 2 weeks after injury but recovered to sham levels by 4 weeks. The contralateral cortex and hippocampus were relatively resistant to changes in neurogranin expression post-injury. Qualitative immunohistochemical assessment corroborated the immunoblot findings. Particularly, the pericontusional cortex and ipsilateral Cornu Ammonis (CA)3 region showed marked reduction in immunoreactivity. PSD-95 demonstrated similar expression patterns to neurogranin in the cortex; however, in the hippocampus, protein expression was increased compared with sham at the 2 and 4 week time points. Our results indicate that CCI lowers neurogranin expression with temporal and regional specificity and that this occurs independently of dendritic loss. Further understanding of the role of neurogranin in synaptic biology after TBI will elucidate pathological mechanisms contributing to cognitive dysfunction.
Collapse
Affiliation(s)
- Sarah Svirsky
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Jeremy Henchir
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Youming Li
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Xiecheng Ma
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Shaun Carlson
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - C. Edward Dixon
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- V.A. Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| |
Collapse
|
36
|
Effect of mild blast-induced TBI on dendritic architecture of the cortex and hippocampus in the mouse. Sci Rep 2020; 10:2206. [PMID: 32042033 PMCID: PMC7010659 DOI: 10.1038/s41598-020-59252-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 01/20/2020] [Indexed: 11/09/2022] Open
Abstract
Traumatic brain injury (TBI) has been designated as a signature injury of modern military conflicts. Blast trauma, in particular, has come to make up a significant portion of the TBIs which are sustained in warzones. Though most TBIs are mild, even mild TBI can induce long term effects, including cognitive and memory deficits. In our study, we utilized a mouse model of mild blast-related TBI (bTBI) to investigate TBI-induced changes within the cortex and hippocampus. We performed rapid Golgi staining on the layer IV and V pyramidal neurons of the parietal cortex and the CA1 basilar tree of the hippocampus and quantified dendritic branching and distribution. We found decreased dendritic branching within both the cortex and hippocampus in injured mice. Within parietal cortex, this decreased branching was most evident within the middle region, while outer and inner regions resembled that of control mice. This study provides important knowledge in the study of how the shockwave associated with a blast explosion impacts different brain regions.
Collapse
|
37
|
Mohamed AZ, Corrigan F, Collins-Praino LE, Plummer SL, Soni N, Nasrallah FA. Evaluating spatiotemporal microstructural alterations following diffuse traumatic brain injury. Neuroimage Clin 2019; 25:102136. [PMID: 31865019 PMCID: PMC6931220 DOI: 10.1016/j.nicl.2019.102136] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Diffuse traumatic brain injury (TBI) is known to lead to microstructural changes within both white and grey matter detected in vivo with diffusion tensor imaging (DTI). Numerous studies have shown alterations in fractional anisotropy (FA) and mean diffusivity (MD) within prominent white matter tracts, but few have linked these to changes within the grey matter with confirmation via histological assessment. This is especially important as alterations in the grey matter may be predictive of long-term functional deficits. METHODS A total of 33 male Sprague Dawley rats underwent severe closed-head TBI. Eight animals underwent tensor-based morphometry (TBM) and DTI at baseline (pre-TBI), 24 hours (24 h), 7, 14, and 30 days post-TBI. Immunohistochemical analysis for the detection of ionised calcium-binding adaptor molecule 1 (IBA1) to assess microglia number and percentage of activated cells, β-amyloid precursor protein (APP) as a marker of axonal injury, and myelin basic protein (MBP) to investigate myelination was performed at each time-point. RESULTS DTI showed significant alterations in FA and RD in numerous white matter tracts including the corpus callosum, internal and external capsule, and optic tract and in the grey-matter in the cortex, thalamus, and hippocampus, with the most significant effects observed at 14 D post-TBI. TBM confirmed volumetric changes within the hippocampus and thalamus. Changes in DTI were in line with significant axonal injury noted at 24 h post-injury via immunohistochemical analysis of APP, with widespread microglial activation seen within prominent white matter tracts and the grey matter, which persisted to 30 D within the hippocampus and thalamus. Microstructural alterations in MBP+ve fibres were also noted within the hippocampus and thalamus, as well as the cortex. CONCLUSION This study confirms the widespread effects of diffuse TBI on white matter tracts which could be detected via DTI and extends these findings to key grey matter regions, with a comprehensive investigation of the whole brain. In particular, the hippocampus and thalamus appear to be vulnerable to ongoing pathology post-TBI, with DTI able to detect these alterations supporting the clinical utility in evaluating these regions post-TBI.
Collapse
Affiliation(s)
- Abdalla Z Mohamed
- Queensland Brain Institute, The University of Queensland, Building 79, Upland Road, Saint Lucia, Brisbane, QLD 4072, Australia
| | - Frances Corrigan
- Head Injury Laboratory, Division of Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Lyndsey E Collins-Praino
- Cognition, Aging and Neurodegenerative Disease Laboratory (CANDL), Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Stephanie L Plummer
- Translational Neuropathology Laboratory, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Neha Soni
- Queensland Brain Institute, The University of Queensland, Building 79, Upland Road, Saint Lucia, Brisbane, QLD 4072, Australia
| | - Fatima A Nasrallah
- Queensland Brain Institute, The University of Queensland, Building 79, Upland Road, Saint Lucia, Brisbane, QLD 4072, Australia.
| |
Collapse
|
38
|
The ameliorative effects of myricetin on neurobehavioral activity, electrophysiology, and biochemical changes in an animal model of traumatic brain injury. LEARNING AND MOTIVATION 2019. [DOI: 10.1016/j.lmot.2019.101597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
39
|
The Recovery of GABAergic Function in the Hippocampus CA1 Region After mTBI. Mol Neurobiol 2019; 57:23-31. [DOI: 10.1007/s12035-019-01753-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 10/26/2022]
|
40
|
Dec K, Łukomska A, Skonieczna-Żydecka K, Kolasa-Wołosiuk A, Tarnowski M, Baranowska-Bosiacka I, Gutowska I. Long-term exposure to fluoride as a factor promoting changes in the expression and activity of cyclooxygenases (COX1 and COX2) in various rat brain structures. Neurotoxicology 2019; 74:81-90. [PMID: 31175943 DOI: 10.1016/j.neuro.2019.06.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 06/03/2019] [Accepted: 06/03/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Sixty percent of the mammalian brain is composed of lipids including arachidonic acid (AA). AA released from cell membranes is metabolised in the cyclooxygenase (COX) pathway to prostanoids - biologically active substances involved in the regulation of many processes including inflammation. It has been shown that long-term exposure to fluoride in pre and neonatal period is dangerous because this element is able to penetrate through the placenta and to cross the blood-brain barrier. Exposure to fluoride during the development affects metabolism and physiology of neurons and glia which results in the impairment of cognitive functions but the exact mechanisms of fluoride neurotoxicity are not clearly defined. OBJECTIVE The aim of this study was to determine whether exposure to fluoride during the development affects COXes activity and the synthesis of prostanoids. MATERIAL AND METHODS Pre- and postnatal toxicity model in Wistar rats was used. Experimental animals received 50 mg/L of NaF in drinking water ad libitum, while control animals received tap water. In cerebral cortex, hippocampus, cerebellum and striatum were measured fluoride concentration, COX1 and COX2 genes expression, immunolocalization of the enzymatic proteins and concentration of PGE2 and TXB2. RESULTS of this study showed statistically significant changes in the concentration of fluoride in brain structures between study group and control animals. Moreover, significant changes in the expression level of COX1 and COX2, and in the concentration of PGE2 and TXB2 were observed. CONCLUSION Exposure to fluoride in the prenatal and neonatal period result in the increase in COX2 activity and increase in PGE2 concentration in rats brain, which may lead to disturbances in central nervous system homeostasis..
Collapse
Affiliation(s)
- Karolina Dec
- The Department of Biochemistry and Human Nutrition, Pomeranian Medical University in Szczecin, Broniewskiego 24 Str., 70-460 Szczecin, Poland
| | - Agnieszka Łukomska
- The Department of Biochemistry and Human Nutrition, Pomeranian Medical University in Szczecin, Broniewskiego 24 Str., 70-460 Szczecin, Poland; Laboratory of Neuroplasticity, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Karolina Skonieczna-Żydecka
- The Department of Biochemistry and Human Nutrition, Pomeranian Medical University in Szczecin, Broniewskiego 24 Str., 70-460 Szczecin, Poland
| | - Agnieszka Kolasa-Wołosiuk
- The Department of Histology and Embryology, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72 av., 70-111 Szczecin, Poland
| | - Maciej Tarnowski
- The Department of Physiology, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72 av., 70-111 Szczecin, Poland
| | - Irena Baranowska-Bosiacka
- The Department of Biochemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72 av., 70-111 Szczecin, Poland
| | - Izabela Gutowska
- The Department of Biochemistry and Human Nutrition, Pomeranian Medical University in Szczecin, Broniewskiego 24 Str., 70-460 Szczecin, Poland.
| |
Collapse
|
41
|
Dief AE, Hassan PS, Hartmut O, Jirikowski GF. Neuronal and glial regeneration after focal cerebral ischemia in rat, an immunohistochemical and electron microscopical study. ALEXANDRIA JOURNAL OF MEDICINE 2019. [DOI: 10.1016/j.ajme.2018.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Abeer E. Dief
- Department of Physiology, University of Alexandria, Egypt
| | | | | | | |
Collapse
|
42
|
Konan LM, Song H, Pentecost G, Fogwe D, Ndam T, Cui J, Johnson CE, Grant D, White T, Chen M, Xia W, Cernak I, DePalma RG, Gu Z. Multi-Focal Neuronal Ultrastructural Abnormalities and Synaptic Alterations in Mice after Low-Intensity Blast Exposure. J Neurotrauma 2019; 36:2117-2128. [PMID: 30667346 DOI: 10.1089/neu.2018.6260] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Service members during military actions or combat training are exposed frequently to primary blast generated by explosive weaponry. The majority of military-related neurotrauma are classified as mild and designated as "invisible injuries" that are prevalent during current conflicts. While the previous experimental blast injury studies using moderate- to high-intensity exposures focused mainly on gross and microscopic neuropathology, our previous studies have shown that low-intensity blast (LIB) exposures resulted in nanoscale subcellular myelin and mitochondrial damages and subsequent behavioral disorders in the absence of gross or detectable cellular damage. In this study, we used transmission electron microscopy to delineate the LIB effects at the ultrastructural level specifically focusing on the neuron perikaryon, axons, and synapses in the cortex and hippocampus of mice at seven and 30 days post-injury (DPI). We found dysmorphic dark neuronal perikaryon and "cytoplasmic aeration" of dendritic processes, as well as increased microtubular fragmentation of the myelinated axons along with biochemically measured elevated tau/phosphorylated tau/Aβ levels. The number of cortical excitatory synapses decreased along with a compensatory increase of the post-synaptic density (PSD) thickness both at seven and 30 DPI, while the amount of hippocampal CA1 synapses increased with the reduced PSD thickness. In addition, we observed a significant increase in protein levels of PSD95 and synaptophysin mainly at seven DPI indicating potential synaptic reorganization. These results demonstrated that a single LIB exposure can lead to ultrastructural brain injury with accompanying multi-focal neuronal organelle alterations. This pre-clinical study provides key insights into disease pathogenesis related to primary blast exposure.
Collapse
Affiliation(s)
- Landry M Konan
- 1 Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri
| | - Hailong Song
- 1 Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri
| | - Genevieve Pentecost
- 1 Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri
| | - Delvise Fogwe
- 1 Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri
| | - Tina Ndam
- 1 Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri
| | - Jiankun Cui
- 1 Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri.,7 Truman VA Hospital Research Service, Columbia, Missouri
| | - Catherine E Johnson
- 2 Department of Mining and Nuclear Engineering, Missouri University of Science and Technology, Rolla, Missouri
| | - DeAna Grant
- 3 Electron Microscopy Core Facility, University of Missouri, Columbia, Missouri
| | - Tommi White
- 3 Electron Microscopy Core Facility, University of Missouri, Columbia, Missouri
| | - Mei Chen
- 4 Bedford VA Medical Center, Bedford, Massachusetts; Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts
| | - Weiming Xia
- 4 Bedford VA Medical Center, Bedford, Massachusetts; Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts
| | - Ibolja Cernak
- 5 STARR-C (Stress, Trauma and Resilience Research Consulting) LLC, Philadelphia, Pennsylvania
| | - Ralph G DePalma
- 6 Norman Rich Department of Surgery, Uniformed University of the Health Sciences, Bethesda, Maryland; Office of Research and Development, Department of Veterans Affairs, Washington, DC
| | - Zezong Gu
- 1 Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri.,7 Truman VA Hospital Research Service, Columbia, Missouri
| |
Collapse
|
43
|
Novgorodov SA, Voltin JR, Wang W, Tomlinson S, Riley CL, Gudz TI. Acid sphingomyelinase deficiency protects mitochondria and improves function recovery after brain injury. J Lipid Res 2019; 60:609-623. [PMID: 30662008 PMCID: PMC6399498 DOI: 10.1194/jlr.m091132] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/11/2019] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the leading causes of disability worldwide and a prominent risk factor for neurodegenerative diseases. The expansion of nervous tissue damage after the initial trauma involves a multifactorial cascade of events, including excitotoxicity, oxidative stress, inflammation, and deregulation of sphingolipid metabolism that further mitochondrial dysfunction and secondary brain damage. Here, we show that a posttranscriptional activation of an acid sphingomyelinase (ASM), a key enzyme of the sphingolipid recycling pathway, resulted in a selective increase of sphingosine in mitochondria during the first week post-TBI that was accompanied by reduced activity of mitochondrial cytochrome oxidase and activation of the Nod-like receptor protein 3 inflammasome. TBI-induced mitochondrial abnormalities were rescued in the brains of ASM KO mice, which demonstrated improved behavioral deficit recovery compared with WT mice. Furthermore, an elevated autophagy in an ASM-deficient brain at the baseline and during the development of secondary brain injury seems to foster the preservation of mitochondria and brain function after TBI. Of note, ASM deficiency attenuated the early stages of reactive astrogliosis progression in an injured brain. These findings highlight the crucial role of ASM in governing mitochondrial dysfunction and brain-function impairment, emphasizing the importance of sphingolipids in the neuroinflammatory response to TBI.
Collapse
Affiliation(s)
- Sergei A Novgorodov
- Departments of Neuroscience Medical University of South Carolina, Charleston, SC 29425
| | - Joshua R Voltin
- Departments of Neuroscience Medical University of South Carolina, Charleston, SC 29425
| | - Wenxue Wang
- Microbiology and Immunology Medical University of South Carolina, Charleston, SC 29425
| | - Stephen Tomlinson
- Microbiology and Immunology Medical University of South Carolina, Charleston, SC 29425
| | | | - Tatyana I Gudz
- Departments of Neuroscience Medical University of South Carolina, Charleston, SC 29425
- Ralph H. Johnson Veterans Affairs Medical Center Charleston, SC 29401
| |
Collapse
|
44
|
Oliver JM, Anzalone AJ, Turner SM. Protection Before Impact: the Potential Neuroprotective Role of Nutritional Supplementation in Sports-Related Head Trauma. Sports Med 2018; 48:39-52. [PMID: 29368186 PMCID: PMC5790849 DOI: 10.1007/s40279-017-0847-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Even in the presence of underreporting, sports-related concussions/mild traumatic brain injuries (mTBI) are on the rise. In the absence of proper diagnosis, an athlete may return to play prior to full recovery, increasing the risk of second-impact syndrome or protracted symptoms. Recent evidence has demonstrated that sub-concussive impacts, those sustained routinely in practice and competition, result in a quantifiable pathophysiological response and the accumulation of both concussive and sub-concussive impacts sustained over a lifetime of sports participation may lead to long-term neurological impairments and an increased risk of developing neurodegenerative diseases. The pathophysiological, neurometabolic, and neurochemical cascade that initiates subsequent to the injury is complex and involves multiple mechanisms. While pharmaceutical treatments may target one mechanism, specific nutrients and nutraceuticals have been discovered to impact several pathways, presenting a broader approach. Several studies have demonstrated the neuroprotective effect of nutritional supplementation in the treatment of mTBI. However, given that many concussions go unreported and sub-concussive impacts result in a pathophysiological response that, too, may contribute to long-term brain health, protection prior to impact is warranted. This review discusses the current literature regarding the role of nutritional supplements that, when provided before mTBI and traumatic brain injury, may provide neurological protection.
Collapse
Affiliation(s)
- Jonathan M Oliver
- Sports Concussion Research Group, Department of Kinesiology, Texas Christian University (TCU), Box 297730, Fort Worth, TX, 76129, USA.
| | - Anthony J Anzalone
- Sports Concussion Research Group, Department of Kinesiology, Texas Christian University (TCU), Box 297730, Fort Worth, TX, 76129, USA
| | - Stephanie M Turner
- Sports Concussion Research Group, Department of Kinesiology, Texas Christian University (TCU), Box 297730, Fort Worth, TX, 76129, USA
| |
Collapse
|
45
|
Arulsamy A, Teng J, Colton H, Corrigan F, Collins-Praino L. Evaluation of early chronic functional outcomes and their relationship to pre-frontal cortex and hippocampal pathology following moderate-severe traumatic brain injury. Behav Brain Res 2018; 348:127-138. [DOI: 10.1016/j.bbr.2018.04.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/20/2018] [Accepted: 04/06/2018] [Indexed: 01/02/2023]
|
46
|
Thau-Zuchman O, Gomes RN, Dyall SC, Davies M, Priestley JV, Groenendijk M, De Wilde MC, Tremoleda JL, Michael-Titus AT. Brain Phospholipid Precursors Administered Post-Injury Reduce Tissue Damage and Improve Neurological Outcome in Experimental Traumatic Brain Injury. J Neurotrauma 2018; 36:25-42. [PMID: 29768974 PMCID: PMC6306688 DOI: 10.1089/neu.2017.5579] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Traumatic brain injury (TBI) leads to cellular loss, destabilization of membranes, disruption of synapses and altered brain connectivity, and increased risk of neurodegenerative disease. A significant and long-lasting decrease in phospholipids (PLs), essential membrane constituents, has recently been reported in plasma and brain tissue, in human and experimental TBI. We hypothesized that supporting PL synthesis post-injury could improve outcome post-TBI. We tested this hypothesis using a multi-nutrient combination designed to support the biosynthesis of PLs and available for clinical use. The multi-nutrient, Fortasyn® Connect (FC), contains polyunsaturated omega-3 fatty acids, choline, uridine, vitamins, cofactors required for PL biosynthesis, and has been shown to have significant beneficial effects in early Alzheimer's disease. Male C57BL/6 mice received a controlled cortical impact injury and then were fed a control diet or a diet enriched with FC for 70 days. FC led to a significantly improved sensorimotor outcome and cognition, reduced lesion size and oligodendrocyte loss, and it restored myelin. It reversed the loss of the synaptic protein synaptophysin and decreased levels of the axon growth inhibitor, Nogo-A, thus creating a permissive environment. It decreased microglia activation and the rise in ß-amyloid precursor protein and restored the depressed neurogenesis. The effects of this medical multi-nutrient suggest that support of PL biosynthesis post-TBI, a new treatment paradigm, has significant therapeutic potential in this neurological condition for which there is no satisfactory treatment. The multi-nutrient tested has been used in dementia patients and is safe and well tolerated, which would enable rapid clinical exploration in TBI.
Collapse
Affiliation(s)
- Orli Thau-Zuchman
- 1 Centre for Neuroscience and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Rita N Gomes
- 1 Centre for Neuroscience and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Simon C Dyall
- 3 Bournemouth University, Royal London House, Bournemouth, United Kingdom
| | - Meirion Davies
- 1 Centre for Neuroscience and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - John V Priestley
- 1 Centre for Neuroscience and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Martine Groenendijk
- 2 Nutricia Research-Nutricia Advanced Medical Nutrition, Utrecht, The Netherlands
| | - Martijn C De Wilde
- 2 Nutricia Research-Nutricia Advanced Medical Nutrition, Utrecht, The Netherlands
| | - Jordi L Tremoleda
- 1 Centre for Neuroscience and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Adina T Michael-Titus
- 1 Centre for Neuroscience and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
47
|
Profound deficits in hippocampal synaptic plasticity after traumatic brain injury and seizure is ameliorated by prophylactic levetiracetam. Oncotarget 2018; 9:11515-11527. [PMID: 29545916 PMCID: PMC5837755 DOI: 10.18632/oncotarget.23923] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/29/2017] [Indexed: 11/25/2022] Open
Abstract
Aim To determine the precise effects of post-traumatic seizure activity on hippocampal processes, we induced seizures at various intervals after traumatic brain injury (TBI) and analyzed plasticity at CA1 Schaffer collateral synapses. Material and Methods Rats were initially separated into two groups; one exposed solely to fluid percussion injury (FPI) at 2 Psi and the other only receiving kainic acid (KA)-induced seizures without FPI. Electrophysiological (ePhys) studies including paired-pulse stimulation for short-term presynaptic plasticity and long-term potentiation (LTP) of CA1 Schaffer collateral synapses of the hippocampus for post-synaptic function survey were followed at post-event 1 hour, 3 and 7 days respectively. Additional rats were exposed to three seizures at weekly intervals starting 1 week or 2 weeks after TBI and compared with seizures without TBI, TBI without seizures, and uninjured animals. An additional group placed under the same control variables were treated with levetiracetam prior to seizure induction. The ePhys studies related to post-TBI induced seizures were also followed in these additional groups. Results Seizures affected the short- and long-term synaptic plasticity of the hippocampal CA3-CA1 pathway. FPI itself suppressed LTP and field excitatory post synaptic potentials (fEPSP) in the CA1 Schaffer collateral synapses; KA-induced seizures that followed FPI further suppressed synaptic plasticity. The impairments in both short-term presynaptic and long-term plasticity were worse in the rats in which early post-TBI seizures were induced than those in which later post-TBI seizures were induced. Finally, prophylactic infusion of levetiracetam for one week after FPI reduced the synaptic plasticity deficits in early post-TBI seizure animals. Conclusion Our data indicates that synaptic plasticity (i.e., both presynaptic and postsynaptic) suppression occurs in TBI followed by a seizure and that the interval between the TBI and seizure is an important factor in the severity of the resulting deficits. Furthermore, the infusion of prophylactic levetiracetam could partially reverse the suppression of synaptic plasticity.
Collapse
|
48
|
Carlson SW, Henchir J, Dixon CE. Lateral Fluid Percussion Injury Impairs Hippocampal Synaptic Soluble N-Ethylmaleimide Sensitive Factor Attachment Protein Receptor Complex Formation. Front Neurol 2017; 8:532. [PMID: 29067000 PMCID: PMC5641299 DOI: 10.3389/fneur.2017.00532] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 09/25/2017] [Indexed: 01/02/2023] Open
Abstract
Traumatic brain injury (TBI) and the activation of secondary injury mechanisms have been linked to impaired cognitive function, which, as observed in TBI patients and animal models, can persist for months and years following the initial injury. Impairments in neurotransmission have been well documented in experimental models of TBI, but the mechanisms underlying this dysfunction are poorly understood. Formation of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex facilitates vesicular docking and neurotransmitter release in the synaptic cleft. Published studies highlight a direct link between reduced SNARE complex formation and impairments in neurotransmitter release. While alterations in the SNARE complex have been described following severe focal TBI, it is not known if deficits in SNARE complex formation manifest in a model with reduced severity. We hypothesized that lateral fluid percussion injury (lFPI) reduces the abundance of SNARE proteins, impairs SNARE complex formation, and contributes to impaired neurobehavioral function. To this end, rats were subjected to lFPI or sham injury and tested for acute motor performance and cognitive function at 3 weeks post-injury. lFPI resulted in motor impairment between 1 and 5 days post-injury. Spatial acquisition and spatial memory, as assessed by the Morris water maze, were significantly impaired at 3 weeks after lFPI. To examine the effect of lFPI on synaptic SNARE complex formation in the injured hippocampus, a separate cohort of rats was generated and brains processed to evaluate hippocampal synaptosomal-enriched lysates at 1 week post-injury. lFPI resulted in a significant reduction in multiple monomeric SNARE proteins, including VAMP2, and α-synuclein, and SNARE complex abundance. The findings in this study are consistent with our previously published observations suggesting that impairments in hippocampal SNARE complex formation may contribute to neurobehavioral dysfunction associated with TBI.
Collapse
Affiliation(s)
- Shaun W Carlson
- Department of Neurosurgery, Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States.,V.A. Pittsburgh Healthcare System, Pittsburgh, PA, United States
| | - Jeremy Henchir
- Department of Neurosurgery, Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States.,V.A. Pittsburgh Healthcare System, Pittsburgh, PA, United States
| | - C Edward Dixon
- Department of Neurosurgery, Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States.,V.A. Pittsburgh Healthcare System, Pittsburgh, PA, United States
| |
Collapse
|
49
|
Saykally JN, Ratliff WA, Keeley KL, Pick CG, Mervis RF, Citron BA. Repetitive Mild Closed Head Injury Alters Protein Expression and Dendritic Complexity in a Mouse Model. J Neurotrauma 2017; 35:139-148. [PMID: 28701108 DOI: 10.1089/neu.2017.5070] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Worldwide head injuries are a growing problem. In the United States alone, 1.7 million people suffer a head injury each year. While most of these injuries are mild, head injury sufferers still sustain symptoms that can have major medical and economical impacts. Moreover, repetitive mild head injuries, like those observed in active military personnel and athletes, have demonstrated a more severe and long-term set of consequences. In an effort to better understand the delayed pathological changes following multiple mild head injuries, we used a mouse model of mild closed head injury (with no motor deficits observed by rotarod testing) and measured dendritic complexity at 30 days after injury and potentially related factors up to 60 days post-injury. We found an increase in TDP-43 protein at 60 days post-injury in the hippocampus and a decrease in autophagy factors three days post-injury. Alterations in dendritic complexity were neuronal subtype and location specific. Measurements of neurotropic factors suggest that an increase in complexity in the cortex may be a consequence of neuronal loss of the less connected neurons.
Collapse
Affiliation(s)
- Jessica N Saykally
- 1 Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System , Bay Pines, Florida.,2 Department of Molecular Medicine, University of South Florida College of Medicine , Tampa, Florida
| | - Whitney A Ratliff
- 1 Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System , Bay Pines, Florida.,2 Department of Molecular Medicine, University of South Florida College of Medicine , Tampa, Florida
| | - Kristen L Keeley
- 1 Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System , Bay Pines, Florida.,2 Department of Molecular Medicine, University of South Florida College of Medicine , Tampa, Florida
| | - Chaim G Pick
- 3 Department of Anatomy and Anthropology, Sackler School of Medicine, Tel Aviv University , Tel Aviv, Israel
| | - Ronald F Mervis
- 4 NeuroStructural Research Laboratories, Inc. , Tampa, Florida.,5 Center for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine , Tampa, Florida
| | - Bruce A Citron
- 1 Laboratory of Molecular Biology, Research and Development 151, Bay Pines VA Healthcare System , Bay Pines, Florida.,2 Department of Molecular Medicine, University of South Florida College of Medicine , Tampa, Florida
| |
Collapse
|
50
|
Dec K, Łukomska A, Maciejewska D, Jakubczyk K, Baranowska-Bosiacka I, Chlubek D, Wąsik A, Gutowska I. The Influence of Fluorine on the Disturbances of Homeostasis in the Central Nervous System. Biol Trace Elem Res 2017; 177:224-234. [PMID: 27787813 PMCID: PMC5418325 DOI: 10.1007/s12011-016-0871-4] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 10/11/2016] [Indexed: 01/21/2023]
Abstract
Fluorides occur naturally in the environment, the daily exposure of human organism to fluorine mainly depends on the intake of this element with drinking water and it is connected with the geographical region. In some countries, we can observe the endemic fluorosis-the damage of hard and soft tissues caused by the excessive intake of fluorine. Recent studies showed that fluorine is toxic to the central nervous system (CNS). There are several known mechanisms which lead to structural brain damage caused by the excessive intake of fluorine. This element is able to cross the blood-brain barrier, and it accumulates in neurons affecting cytological changes, cell activity and ion transport (e.g. chlorine transport). Additionally, fluorine changes the concentration of non-enzymatic advanced glycation end products (AGEs), the metabolism of neurotransmitters (influencing mainly glutamatergic neurotransmission) and the energy metabolism of neurons by the impaired glucose transporter-GLUT1. It can also change activity and lead to dysfunction of important proteins which are part of the respiratory chain. Fluorine also affects oxidative stress, glial activation and inflammation in the CNS which leads to neurodegeneration. All of those changes lead to abnormal cell differentiation and the activation of apoptosis through the changes in the expression of neural cell adhesion molecules (NCAM), glial fibrillary acidic protein (GFAP), brain-derived neurotrophic factor (BDNF) and MAP kinases. Excessive exposure to this element can cause harmful effects such as permanent damage of all brain structures, impaired learning ability, memory dysfunction and behavioural problems. This paper provides an overview of the fluoride neurotoxicity in juveniles and adults.
Collapse
Affiliation(s)
- K Dec
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University in Szczecin, Broniewskiego street 24, 70-406, Szczecin, Poland
| | - A Łukomska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University in Szczecin, Broniewskiego street 24, 70-406, Szczecin, Poland
| | - D Maciejewska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University in Szczecin, Broniewskiego street 24, 70-406, Szczecin, Poland
| | - K Jakubczyk
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University in Szczecin, Broniewskiego street 24, 70-406, Szczecin, Poland
| | - I Baranowska-Bosiacka
- Department of Biochemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72 av., 71-111, Szczecin, Poland
| | - D Chlubek
- Department of Biochemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72 av., 71-111, Szczecin, Poland
| | - A Wąsik
- Institute of Pharmacology, Polish Academy of Sciences, Department of Neurochemistry, Smętna street 12, 31-343, Kraków, Poland
| | - I Gutowska
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University in Szczecin, Broniewskiego street 24, 70-406, Szczecin, Poland.
| |
Collapse
|