101
|
Uddin O, Studlack PE, Parihar S, Keledjian K, Cruz A, Farooq T, Shin N, Gerzanich V, Simard JM, Keller A. Chronic pain after blast-induced traumatic brain injury in awake rats. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2019; 6:100030. [PMID: 31223145 PMCID: PMC6565615 DOI: 10.1016/j.ynpai.2019.100030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/14/2019] [Accepted: 04/01/2019] [Indexed: 12/14/2022]
Abstract
Explosive blast-induced traumatic brain injury (blast-TBI) in military personnel is a leading cause of injury and persistent neurological abnormalities, including chronic pain. We previously demonstrated that chronic pain after spinal cord injury results from central sensitization in the posterior thalamus (PO). The presence of persistent headaches and back pain in veterans with blast-TBI suggests a similar involvement of thalamic sensitization. Here, we tested the hypothesis that pain after blast-TBI is associated with abnormal increases in activity of neurons in PO thalamus. We developed a novel model with two unique features: (1) blast-TBI was performed in awake, un-anesthetized rats, to simulate the human experience and to eliminate confounds of anesthesia and surgery inherent in other models; (2) only the cranium, rather than the entire body, was exposed to a collimated blast wave, with the blast wave striking the posterior cranium in the region of the occipital crest and foramen magnum. Three weeks after blast-TBI, rats developed persistent, ongoing spontaneous pain. Contrary to our hypothesis, we found no significant differences in the activity of PO neurons, or of neurons in the spinal trigeminal nucleus. There were also no significant changes in gliosis in either of these structures. This novel model will allow future studies on the pathophysiology of chronic pain after blast-TBI.
Collapse
Affiliation(s)
- Olivia Uddin
- Department of Anatomy and Neurobiology and Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD, USA
| | - Paige E. Studlack
- Department of Anatomy and Neurobiology and Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD, USA
| | - Saitu Parihar
- Department of Anatomy and Neurobiology and Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD, USA
| | - Kaspar Keledjian
- Department of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF 634B, Baltimore, MD, USA
| | - Alexis Cruz
- Department of Anatomy and Neurobiology and Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD, USA
| | - Tayyiaba Farooq
- Department of Anatomy and Neurobiology and Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD, USA
| | - Naomi Shin
- Department of Anatomy and Neurobiology and Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF 634B, Baltimore, MD, USA
| | - J. Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF 634B, Baltimore, MD, USA
- Department of Pathology, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD, USA
- Department of Physiology, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD, USA
| | - Asaf Keller
- Department of Anatomy and Neurobiology and Program in Neuroscience, University of Maryland School of Medicine, 20 Penn St, HSF-II S251, Baltimore, MD, USA
| |
Collapse
|
102
|
Pham L, Shultz SR, Kim HA, Brady RD, Wortman RC, Genders SG, Hale MW, O'Shea RD, Djouma E, van den Buuse M, Church JE, Christie BR, Drummond GR, Sobey CG, McDonald SJ. Mild Closed-Head Injury in Conscious Rats Causes Transient Neurobehavioral and Glial Disturbances: A Novel Experimental Model of Concussion. J Neurotrauma 2019; 36:2260-2271. [DOI: 10.1089/neu.2018.6169] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Louise Pham
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, Australia
| | - Sandy R. Shultz
- Department Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Hyun Ah Kim
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, Australia
| | - Rhys D. Brady
- Department Neuroscience, Monash University, Melbourne, Australia
- Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Ryan C. Wortman
- Department Neuroscience, Monash University, Melbourne, Australia
| | - Shannyn G. Genders
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, Australia
| | - Matthew W. Hale
- Department of Psychology and Counseling, La Trobe University, Bundoora, Australia
| | - Ross D. O'Shea
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, Australia
| | - Elvan Djouma
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, Australia
| | - Maarten van den Buuse
- Department of Psychology and Counseling, La Trobe University, Bundoora, Australia
- Department of Pharmacology, University of Melbourne, Melbourne, Australia
- The College of Public Health, Medical, and Veterinary Sciences, James Cook University, Queensland, Australia
| | - Jarrod E. Church
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, Australia
| | - Brian R. Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Grant R. Drummond
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, Australia
| | - Christopher G. Sobey
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, Australia
| | - Stuart J. McDonald
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, Australia
- Department Neuroscience, Monash University, Melbourne, Australia
| |
Collapse
|
103
|
Abstract
The underlying mechanisms that result in neurophysiological changes and cognitive sequelae in the context of repetitive mild traumatic brain injury (rmTBI) remain poorly understood. Animal models provide a unique opportunity to examine cellular and molecular responses using histological assessment, which can give important insights on the neurophysiological changes associated with the evolution of brain injury. To better understand the potential cumulative effects of multiple concussions, the focus of animal models is shifting from single to repetitive head impacts. With a growing body of literature on this subject, a review and discussion of current findings is valuable to better understand the neuropathology associated with rmTBI, to evaluate the current state of the field, and to guide future research efforts. Despite variability in experimental settings, existing animal models of rmTBI have contributed to our understanding of the underlying mechanisms following repeat concussion. However, how to reconcile the various impact methods remains one of the major challenges in the field today.
Collapse
Affiliation(s)
- Wouter S Hoogenboom
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA; Department of Clinical Investigation, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA.
| | - Craig A Branch
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA; Department of Physiology and Biophysics, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA; Department of Radiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA.
| | - Michael L Lipton
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA; Department of Radiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA; Departments of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA; The Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA.
| |
Collapse
|
104
|
Hiskens MI, Angoa-Pérez M, Schneiders AG, Vella RK, Fenning AS. Modeling sports-related mild traumatic brain injury in animals-A systematic review. J Neurosci Res 2019; 97:1194-1222. [PMID: 31135069 DOI: 10.1002/jnr.24472] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/22/2019] [Accepted: 05/07/2019] [Indexed: 12/14/2022]
Abstract
Sports-related head trauma has emerged as an important public health issue, as mild traumatic brain injuries (mTBIs) may result in neurodegenerative disorders such as chronic traumatic encephalopathy (CTE). Research into mTBI and CTE pathophysiology are difficult to undertake in athletes, with observational trials and post-mortem analysis the current mainstays. Thus, animal models play an important role in the study of mTBI, however, traditional animal models have focused on acute, severe injuries rather than the more typical mTBI's seen in sport injuries. Recently, a number of animal models have been developed that are both appropriately scaled and biomechanically relevant to the forces sustained by athletes. This review aimed to examine the literature for variables included in these animal models, and the resulting neurotrauma as evidenced by pathology and behavioral deficits. A systematic search of the literature was performed in multiple electronic databases. The inclusion criteria required mimicry of athlete mTBI conditions: freedom of head movement, lack of surgical alteration of the skull, and application of direct contact force. Studies were analyzed for variables including apparatus design features (impact force, change in animal head velocity, and kinetic energy transfer to the head), demonstrated pathology (phosphorylated tau, TDP-43 aggregation, diffuse axonal injury, gliosis, cytokine inflammation response, and genetic integrity), and behavioral changes. These studies suggested that appropriate animal models can assist in understanding the pathological and functional outcomes of athlete mTBI, and could be used as a platform for future studies of diagnostic/prognostic markers and in the development of treatment interventions.
Collapse
Affiliation(s)
- Matthew I Hiskens
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, Australia
| | - Mariana Angoa-Pérez
- Research & Development Service, John D. Dingell VA Medical Center, Detroit, Michigan.,Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan
| | - Anthony G Schneiders
- School of Health, Medical and Applied Sciences, Central Queensland University, Branyan, Australia
| | - Rebecca K Vella
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, Australia
| | - Andrew S Fenning
- School of Health, Medical and Applied Sciences, Central Queensland University, Rockhampton, Australia
| |
Collapse
|
105
|
Koletar MM, Dorr A, Brown ME, McLaurin J, Stefanovic B. Refinement of a chronic cranial window implant in the rat for longitudinal in vivo two-photon fluorescence microscopy of neurovascular function. Sci Rep 2019; 9:5499. [PMID: 30940849 PMCID: PMC6445076 DOI: 10.1038/s41598-019-41966-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 03/19/2019] [Indexed: 12/13/2022] Open
Abstract
Longitudinal studies using two–photon fluorescence microscopy (TPFM) are critical for facilitating cellular scale imaging of brain morphology and function. Studies have been conducted in the mouse due to their relatively higher transparency and long term patency of a chronic cranial window. Increasing availability of transgenic rat models, and the range of established behavioural paradigms, necessitates development of a chronic preparation for the rat. However, surgical craniotomies in the rat present challenges due to craniotomy closure by wound healing and diminished image quality due to inflammation, restricting most rat TPFM experiments to acute preparations. Long-term patency is enabled by employing sterile surgical technique, minimization of trauma with precise tissue handling during surgery, judicious selection of the size and placement of the craniotomy, diligent monitoring of animal physiology and support throughout the surgery, and modification of the home cage for long-term preservation of cranial implants. Immunohistochemical analysis employing the glial fibrillary acidic protein (GFAP) and ionized calcium-binding adaptor molecule-1 (Iba-1) showed activation and recruitment of astrocytes and microglia/macrophages directly inferior to the cranial window at one week after surgery, with more diffuse response in deeper cortical layers at two weeks, and amelioration around four weeks post craniotomy. TPFM was conducted up to 14 weeks post craniotomy, reaching cortical depths of 400 µm to 600 µm at most time-points. The rate of signal decay with increasing depth and maximum cortical depth attained had greater variation between individual rats at a single time-point than within a rat across time.
Collapse
Affiliation(s)
- Margaret M Koletar
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, Ontario, M4N 3M5, Canada.
| | - Adrienne Dorr
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, Ontario, M4N 3M5, Canada
| | - Mary E Brown
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, Ontario, M4N 3M5, Canada
| | - JoAnne McLaurin
- Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, Ontario, M4N 3M5, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A1, Canada
| | - Bojana Stefanovic
- Department of Medical Biophysics, University of Toronto, 610 University Avenue, Toronto, Ontario, M5G 2M9, Canada.,Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, Ontario, M4N 3M5, Canada
| |
Collapse
|
106
|
Subacute to chronic Alzheimer-like alterations after controlled cortical impact in human tau transgenic mice. Sci Rep 2019; 9:3789. [PMID: 30846870 PMCID: PMC6405988 DOI: 10.1038/s41598-019-40678-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 02/21/2019] [Indexed: 12/14/2022] Open
Abstract
Repetitive traumatic brain injury (TBI) has been linked to late life development of chronic traumatic encephalopathy (CTE), a neurodegenerative disorder histopathologically characterized by perivascular tangles of hyperphosphorylated tau at the depth of sulci to later widespread neurofibrillary pathology. Although tau hyperphosphorylation and neurofibrillary-like pathology have been observed in the brain of transgenic mice overexpressing human tau with aggregation-prone mutation after TBI, they have not been consistently recapitulated in rodents expressing wild-type tau only. Here, we characterized Alzheimer-like alterations behaviorally, biochemically and immunohistochemically 6 weeks and 7 months after unilateral mild-to-moderate controlled cortical impact (CCI) in 5–7-month-old Tg/htau mice, which express all six isoforms of non-mutated human tau in a mouse tau null background. We detected hyperphosphorylation of tau at multiple sites in ipsilateral hippocampus 6 weeks but not 7 months after CCI. However, neuronal accumulation of AT8 positive phospho-tau was sustained in the chronic phase, in parallel to prolonged astrogliosis, and decreased neural and synaptic markers. The mice with CCI also exhibited cognitive and locomotor impairment. These results indicate subacute to chronic Alzheimer-like alterations after CCI in Tg/htau mice. This is the first known study providing insight into the role of CCI in Alzheimer-like brain alterations in young adult mice expressing only non-mutated human tau.
Collapse
|
107
|
Chao H, Lin C, Zuo Q, Liu Y, Xiao M, Xu X, Li Z, Bao Z, Chen H, You Y, Kochanek PM, Yin H, Liu N, Kagan VE, Bayır H, Ji J. Cardiolipin-Dependent Mitophagy Guides Outcome after Traumatic Brain Injury. J Neurosci 2019; 39:1930-1943. [PMID: 30626699 PMCID: PMC6407296 DOI: 10.1523/jneurosci.3415-17.2018] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 11/21/2018] [Accepted: 12/28/2018] [Indexed: 01/14/2023] Open
Abstract
Mitochondrial energy production is essential for normal brain function. Traumatic brain injury (TBI) increases brain energy demands, results in the activation of mitochondrial respiration, associated with enhanced generation of reactive oxygen species. This chain of events triggers neuronal apoptosis via oxidation of a mitochondria-specific phospholipid, cardiolipin (CL). One pathway through which cells can avoid apoptosis is via elimination of damaged mitochondria by mitophagy. Previously, we showed that externalization of CL to the mitochondrial surface acts as an elimination signal in cells. Whether CL-mediated mitophagy occurs in vivo or its significance in the disease processes are not known. In this study, we showed that TBI leads to increased mitophagy in the human brain, which was also detected using TBI models in male rats. Knockdown of CL synthase, responsible for de novo synthesis of CL, or phospholipid scramblase-3, responsible for CL translocation to the outer mitochondrial membrane, significantly decreased TBI-induced mitophagy. Inhibition of mitochondrial clearance by 3-methyladenine, mdivi-1, or phospholipid scramblase-3 knockdown after TBI led to a worse outcome, suggesting that mitophagy is beneficial. Together, our findings indicate that TBI-induced mitophagy is an endogenous neuroprotective process that is directed by CL, which marks damaged mitochondria for elimination, thereby limiting neuronal death and behavioral deficits.SIGNIFICANCE STATEMENT Traumatic brain injury (TBI) increases energy demands leading to activation of mitochondrial respiration associated with enhanced generation of reactive oxygen species and resultant damage to mitochondria. We demonstrate that the complete elimination of irreparably damaged organelles via mitophagy is activated as an early response to TBI. This response includes translocation of mitochondria phospholipid cardiolipin from the inner membrane to the outer membrane where externalized cardiolipin mediates targeted protein light chain 3-mediated autophagy of damaged mitochondria. Our data on targeting phospholipid scramblase and cardiolipin synthase in genetically manipulated cells and animals strongly support the essential role of cardiolipin externalization mechanisms in the endogenous reparative plasticity of injured brain cells. Furthermore, successful execution and completion of mitophagy is beneficial in the context of preservation of cognitive functions after TBI.
Collapse
Affiliation(s)
- Honglu Chao
- Departments of Neurosurgery and
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| | | | - Qiang Zuo
- Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | | | - Mengqing Xiao
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS), Shanghai 200031, China
- University of the Chinese Academy of Sciences, CAS, Beijing 100049, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
| | | | | | | | - Huimei Chen
- Department of Medical Genetics, Nanjing University School of Medicine, Nanjing 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210029, China
| | | | - Patrick M Kochanek
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| | - Huiyong Yin
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS), Shanghai 200031, China
- University of the Chinese Academy of Sciences, CAS, Beijing 100049, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
- Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing 100022, China
| | | | - Valerian E Kagan
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health
- Laboratory of Navigational Redox Lipidomics and Department of Human Pathology, IM Sechenov Moscow State Medical University, Moscow 119991, Russian Federation, and
| | - Hülya Bayır
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health,
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
- Children's Neuroscience Institute, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania 15224
| | - Jing Ji
- Departments of Neurosurgery and
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| |
Collapse
|
108
|
Andrade P, Banuelos-Cabrera I, Lapinlampi N, Paananen T, Ciszek R, Ndode-Ekane XE, Pitkänen A. Acute Non-Convulsive Status Epilepticus after Experimental Traumatic Brain Injury in Rats. J Neurotrauma 2019; 36:1890-1907. [PMID: 30543155 DOI: 10.1089/neu.2018.6107] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Severe traumatic brain injury (TBI) induces seizures or status epilepticus (SE) in 20-30% of patients during the acute phase. We hypothesized that severe TBI induced with lateral fluid-percussion injury (FPI) triggers post-impact SE. Adult Sprague-Dawley male rats were anesthetized with isoflurane and randomized into the sham-operated experimental control or lateral FPI-induced severe TBI groups. Electrodes were implanted right after impact or sham-operation, then video-electroencephalogram (EEG) monitoring was started. In addition, video-EEG was recorded from naïve rats. During the first 72 h post-TBI, injured rats had seizures that were intermingled with other epileptiform EEG patterns typical to non-convulsive SE, including occipital intermittent rhythmic delta activity, lateralized or generalized periodic discharges, spike-and-wave complexes, poly-spikes, poly-spike-and-wave complexes, generalized continuous spiking, burst suppression, or suppression. Almost all (98%) of the electrographic seizures were recorded during 0-72 h post-TBI (23.2 ± 17.4 seizures/rat). Mean latency from the impact to the first electrographic seizure was 18.4 ± 15.1 h. Mean seizure duration was 86 ± 57 sec. Analysis of high-resolution videos indicated that only 41% of electrographic seizures associated with behavioral abnormalities, which were typically subtle (Racine scale 1-2). Fifty-nine percent of electrographic seizures did not show any behavioral manifestations. In most of the rats, epileptiform EEG patterns began to decay spontaneously on Days 5-6 after TBI. Interestingly, also a few sham-operated and naïve rats had post-operation seizures, which were not associated with EEG background patterns typical to non-convulsive SE seen in TBI rats. To summarize, our data show that lateral FPI-induced TBI results in non-convulsive SE with subtle behavioral manifestations; this explains why it has remained undiagnosed until now. The lateral FPI model provides a novel platform for assessing the mechanisms of acute symptomatic non-convulsive SE and for testing treatments to prevent post-injury SE in a clinically relevant context.
Collapse
Affiliation(s)
- Pedro Andrade
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Ivette Banuelos-Cabrera
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Niina Lapinlampi
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tomi Paananen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Robert Ciszek
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | | - Asla Pitkänen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
109
|
Corne R, Leconte C, Ouradou M, Fassina V, Zhu Y, Déou E, Besson V, Plotkine M, Marchand-Leroux C, Mongeau R. Spontaneous resurgence of conditioned fear weeks after successful extinction in brain injured mice. Prog Neuropsychopharmacol Biol Psychiatry 2019; 88:276-286. [PMID: 30096331 DOI: 10.1016/j.pnpbp.2018.07.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/28/2018] [Accepted: 07/29/2018] [Indexed: 12/25/2022]
Abstract
Mild traumatic brain injury (TBI) is a major risk factor for post-traumatic stress disorder (PTSD), and both disorders share common symptoms and neurobiological defects. Relapse after successful treatment, known as long-term fear resurgence, is common in PTSD patients and a major therapeutic hurdle. We induced a mild focal TBI by controlled cortical impact (CCI) in male C57BL/6 J mice and used fear conditioning to assess PTSD-like behaviors and concomitant alterations in the fear circuitry. We found for the first time that mild TBI, and to a lesser extent sham (craniotomy), mice displayed a spontaneous resurgence of conditioned fear when tested for fear extinction memory recall, despite having effectively acquired and extinguished conditioned fear 6 weeks earlier in the same context. Other characteristic symptoms of PTSD are risk-taking behaviors and cognitive deficits. CCI mice displayed risk-taking behaviors, behavioral inflexibility and reductions in processing speed compared to naïve mice. In conjunction with these changes there were alterations in amygdala morphology 3 months post-trauma, and decreased myelin basic protein density at the primary lesion site and in distant secondary sites such as the hippocampus, thalamus, and amygdala, compared to sham mice. Furthermore, activity-dependent brain-derived neurotrophic factor (BDNF) transcripts were decreased in the prefrontal cortex, a key region for fear extinction consolidation, following fear extinction training in both TBI and, to a lesser extent, sham mice. This study shows for the first time that a mild brain injury can generate a spontaneous resurgence of conditioned fear associated with defective BDNF signalling in the prefrontal cortex, PTSD-like behaviors, and have enduring effects on the brain.
Collapse
Affiliation(s)
- R Corne
- EA4475 - Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France
| | - C Leconte
- EA4475 - Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France
| | - M Ouradou
- EA4475 - Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France
| | - V Fassina
- EA4475 - Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France
| | - Y Zhu
- EA4475 - Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France
| | - E Déou
- EA4475 - Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France
| | - V Besson
- EA4475 - Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France
| | - M Plotkine
- EA4475 - Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France
| | - C Marchand-Leroux
- EA4475 - Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France
| | - R Mongeau
- EA4475 - Pharmacologie de la Circulation Cérébrale, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France.
| |
Collapse
|
110
|
Eles JR, Vazquez AL, Kozai TDY, Cui XT. Meningeal inflammatory response and fibrous tissue remodeling around intracortical implants: An in vivo two-photon imaging study. Biomaterials 2018; 195:111-123. [PMID: 30634095 DOI: 10.1016/j.biomaterials.2018.12.031] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/15/2018] [Accepted: 12/28/2018] [Indexed: 12/21/2022]
Abstract
Meningeal inflammation and encapsulation of neural electrode arrays is a leading cause of device failure, yet little is known about how it develops over time or what triggers it. This work characterizes the dynamic changes of meningeal inflammatory cells and collagen-I in order to understand the meningeal tissue response to neural electrode implantation. We use in vivo two-photon microscopy of CX3CR1-GFP mice over the first month after electrode implantation to quantify changes in inflammatory cell behavior as well as meningeal collagen-I remodeling. We define a migratory window during the first day after electrode implantation hallmarked by robust inflammatory cell migration along electrodes in the meninges as well as cell trafficking through meningeal venules. This migratory window attenuates by 2 days post-implant, but over the next month, the meningeal collagen-I remodels to conform to the surface of the electrode and thickens. This work shows that there are distinct time courses for initial meningeal inflammatory cell infiltration and meningeal collagen-I remodeling. This may indicate a therapeutic window early after implantation for modulation and mitigation of meningeal inflammation.
Collapse
Affiliation(s)
- J R Eles
- Bioengineering, University of Pittsburgh, United States; Center for the Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, United States
| | - A L Vazquez
- Bioengineering, University of Pittsburgh, United States; Center for the Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, United States; Radiology, University of Pittsburgh, United States
| | - T D Y Kozai
- Bioengineering, University of Pittsburgh, United States; Center for the Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, United States; NeuroTech Center of the University of Pittsburgh Brain Institute, United States; Center for Neuroscience, University of Pittsburgh, United States
| | - X T Cui
- Bioengineering, University of Pittsburgh, United States; Center for the Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, United States.
| |
Collapse
|
111
|
Siebold L, Obenaus A, Goyal R. Criteria to define mild, moderate, and severe traumatic brain injury in the mouse controlled cortical impact model. Exp Neurol 2018; 310:48-57. [DOI: 10.1016/j.expneurol.2018.07.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/05/2018] [Accepted: 07/11/2018] [Indexed: 10/28/2022]
|
112
|
Anjum SMM, Käufer C, Hopfengärtner R, Waltl I, Bröer S, Löscher W. Automated quantification of EEG spikes and spike clusters as a new read out in Theiler's virus mouse model of encephalitis-induced epilepsy. Epilepsy Behav 2018; 88:189-204. [PMID: 30292054 DOI: 10.1016/j.yebeh.2018.09.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/13/2018] [Accepted: 09/16/2018] [Indexed: 12/17/2022]
Abstract
Intracerebral infection of C57BL/6 mice with Theiler's murine encephalomyelitis virus (TMEV) replicates many features of viral encephalitis-induced epilepsy in humans, including neuroinflammation, early (insult-associated) and late (spontaneous) seizures, neurodegeneration in the hippocampus, and cognitive and behavioral alterations. Thus, this model may be ideally suited to study mechanisms involved in encephalitis-induced epilepsy as potential targets for epilepsy prevention. However, spontaneous recurrent seizures (SRS) occur too infrequently to be useful as a biomarker of epilepsy, e.g., for drug studies. This prompted us to evaluate whether epileptiform spikes or spike clusters in the cortical electroencephalogram (EEG) may be a useful surrogate of epilepsy in this model. For this purpose, we developed an algorithm that allows efficient and large-scale EEG analysis of early and late seizures, spikes, and spike clusters in the EEG. While 77% of the infected mice exhibited early seizures, late seizures were only observed in 33% of the animals. The clinical characteristics of early and late seizures did not differ except that late generalized convulsive (stage 5) seizures were significantly longer than early stage 5 seizures. Furthermore, the frequency of SRS was much lower than the frequency of early seizures. Continuous (24/7) video-EEG monitoring over several months following infection indicated that the latent period to onset of SRS was 61 (range 16-91) days. Spike and spike clusters were significantly more frequent in infected mice with late seizures than in infected mice without seizures or in mock-infected sham controls. Based on the results of this study, increases in EEG spikes and spike clusters in groups of infected mice may be used as a new readout for studies on antiepileptogenic or disease-modifying drug effects in this model, because the significant increase in average spike counts in mice with late seizures obviously indicates a proepileptogenic alteration.
Collapse
Affiliation(s)
- Syed Muhammad Muneeb Anjum
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Christopher Käufer
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | | | - Inken Waltl
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Sonja Bröer
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
113
|
Wortman RC, Meconi A, Neale KJ, Brady RD, McDonald SJ, Christie BR, Wright DK, Shultz SR. Diffusion MRI abnormalities in adolescent rats given repeated mild traumatic brain injury. Ann Clin Transl Neurol 2018; 5:1588-1598. [PMID: 30564624 PMCID: PMC6292182 DOI: 10.1002/acn3.667] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/12/2018] [Accepted: 09/20/2018] [Indexed: 12/14/2022] Open
Abstract
Objective Mild traumatic brain injury (mTBI) is a serious health concern in the adolescent population. Repeated mTBI may result in more pronounced deficits, and has been associated with long‐term neurological consequences including neurodegeneration. As such, there is a critical need for the development of objective mTBI biomarkers to help guide medical management. Diffusion‐weighted imaging (DWI) is an advanced magnetic resonance imaging (MRI) technique that may detect brain abnormalities after mTBI. Diffusion tensor imaging (DTI) is the most commonly applied DWI method, and initial studies have reported DTI changes in mTBI patients. Furthermore, new DWI methods (e.g., track‐weighted imaging; TWI) are being developed that may also be sensitive to mTBIs, but remain to be comprehensively studied. Methods This study utilized the Awake Closed Head Injury (ACHI) model of mTBI to investigate changes in DTI and TWI following repeated mTBI in adolescent male and female rats. A total of four ACHI impacts, two/day over two consecutive days, were delivered beginning on postnatal day 25. At 1 day and 7 days postinjury, rats were euthanized and brains were collected for DWI analyses. Results Rats given repeated mTBI displayed changes in fractional anisotropy and radial diffusivity (i.e., DTI measures), as well as track density (i.e., TWI). Interpretation These findings are consistent with initial DTI findings in mTBI patients, suggest that TWI may complement DTI, support the utility of DWI measures as biomarkers in mTBI, and further validate the ACHI rat model of mTBI.
Collapse
Affiliation(s)
- Ryan C Wortman
- Department of Neuroscience Central Clinical School Monash University Melbourne Victoria 3004 Australia.,Division of Medical Sciences University of Victoria Victoria BC V8P 5C2 Canada
| | - Alicia Meconi
- Department of Neuroscience Central Clinical School Monash University Melbourne Victoria 3004 Australia
| | - Katie J Neale
- Division of Medical Sciences University of Victoria Victoria BC V8P 5C2 Canada
| | - Rhys D Brady
- Department of Neuroscience Central Clinical School Monash University Melbourne Victoria 3004 Australia
| | - Stuart J McDonald
- Department of Physiology, Anatomy, and Microbiology La Trobe University Bundoora Victoria 3086 Australia
| | - Brian R Christie
- Division of Medical Sciences University of Victoria Victoria BC V8P 5C2 Canada
| | - David K Wright
- Department of Neuroscience Central Clinical School Monash University Melbourne Victoria 3004 Australia.,The Florey Institute of Neuroscience and Mental Health Parkville Victoria 3052 Australia
| | - Sandy R Shultz
- Department of Neuroscience Central Clinical School Monash University Melbourne Victoria 3004 Australia.,Division of Medical Sciences University of Victoria Victoria BC V8P 5C2 Canada.,Department of Medicine The Royal Melbourne Hospital The University of Melbourne Parkville Victoria 3010 Australia
| |
Collapse
|
114
|
Xing G, Ren M, Verma A. Divergent Induction of Branched-Chain Aminotransferases and Phosphorylation of Branched Chain Keto-Acid Dehydrogenase Is a Potential Mechanism Coupling Branched-Chain Keto-Acid-Mediated-Astrocyte Activation to Branched-Chain Amino Acid Depletion-Mediated Cognitive Deficit after Traumatic Brain Injury. J Neurotrauma 2018; 35:2482-2494. [PMID: 29764289 PMCID: PMC6196747 DOI: 10.1089/neu.2017.5496] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Deficient branched-chain amino acids (BCAAs) are implicated in cognitive dysfunction after traumatic brain injury (TBI). The mechanism remains unknown. BCAAs are catabolized by neuron-specific cytosolic and astrocyte-specific mitochondrial branched-chain aminotransferases (BCATc, BCATm) to generate glutamate and branched-chain keto-acids (BCKAs) that are metabolized by the mitochondrial branched-chain keto-acid dehydrogenase (BCKD) whose activity is regulated by its phosphorylation state. BCKD phosphorylation by BCKD kinase (BCKDK) inactivates BCKD and cause neurocognitive dysfunction, whereas dephosphorylation by specific phosphatase restores BCKD activity. Real-time polymerase chain reaction showed rapidly and significantly decreased BCATc messenger RNA (mRNA) levels, but significantly increased BCATm mRNA level post-CCI (controlled cortical impact). BCKD and BCKDK mRNA decreased significantly immediately after CCI-induced TBI (CCI) in the rat. Phosphorylated BCKD proteins (pBCKD) increased significantly in the ipsilateral-CCI hemisphere. Immunohistochemistry revealed significantly increased pBCKD proteins in ipsilateral astrocytes post-CCI. BCKD protein expression is higher in primarily cultured cortical neurons than in astrocytes, whereas pBCKD protein level is higher in astrocytes than in cortical neurons. Transforming growth factor beta treatment (10 μg/mL for 48 h) significantly increased pBCKD protein expression in astrocytes, whereas glutamate treatment (25 μM for 24 h) significantly decreased pBCKD protein in neurons. Because increased pBCKD would lead to increased BCKA accumulation, BCKA-mediated astrocyte activation, cell death, and cognitive dysfunction as found in maple syrup urine disease; thus, TBI may potentially induce cognitive deficit through diverting BCAA from glutamate production in neurons to BCKA production in astrocytes through the pBCKD-dependent mechanism.
Collapse
Affiliation(s)
- Guoqiang Xing
- Department of Radiology and Imaging, Institute of Rehabilitation and Development of Brain Function, The Second Clinical Medical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, China
| | - Ming Ren
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | | |
Collapse
|
115
|
Shaver TK, Ozga JE, Zhu B, Anderson KG, Martens KM, Vonder Haar C. Long-term deficits in risky decision-making after traumatic brain injury on a rat analog of the Iowa gambling task. Brain Res 2018; 1704:103-113. [PMID: 30296430 DOI: 10.1016/j.brainres.2018.10.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/28/2018] [Accepted: 10/04/2018] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) affects 2.8 million people annually in the United States, with significant populations suffering from ongoing cognitive dysfunction. Impairments in decision-making can have major implications for patients and their caregivers, often enduring for years to decades, yet are rarely explored in experimental TBI. In the current study, the Rodent Gambling Task (RGT), an Iowa Gambling Task analog, was used to assess risk-based decision-making and motor impulsivity after TBI. During testing, rats chose between options associated with different probabilities of reinforcement (sucrose) or punishment (timeout). To determine effects of TBI on learned behaviors versus the learning process, rats were trained either before, or after, a bilateral frontal controlled cortical impact TBI, and then assessed for 12 weeks. To evaluate the degree to which monoamine systems, such as dopamine, were affected by TBI, rats were given an amphetamine challenge, and behavior recorded. Injury immediately and chronically decreased optimal decision-making, and biased rats towards both riskier, and safer (but suboptimal) choices, regardless of prior learning history. TBI also increased motor impulsivity across time, reflecting ongoing neural changes. Despite these similarities in trained and acquisition rats, those that learned the task after injury demonstrated reduced effects of amphetamine on optimal decision-making, suggesting a lesser role of monoamines in post-injury learning. Amphetamine also dose-dependently reduced motor impulsivity in injured rats. This study opens up the investigation of psychiatric-like dysfunction in animal models of TBI and tasks such as the RGT will be useful in identifying therapeutics for the chronic post-injury period.
Collapse
Affiliation(s)
- Trinity K Shaver
- Department of Psychology, West Virginia University, Morgantown, WV, USA
| | - Jenny E Ozga
- Department of Psychology, West Virginia University, Morgantown, WV, USA
| | - Binxing Zhu
- Department of Psychology, West Virginia University, Morgantown, WV, USA
| | - Karen G Anderson
- Department of Psychology, West Virginia University, Morgantown, WV, USA
| | - Kris M Martens
- Department of Psychology, West Virginia University, Morgantown, WV, USA
| | - Cole Vonder Haar
- Department of Psychology, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
116
|
Missault S, Anckaerts C, Blockx I, Deleye S, Van Dam D, Barriche N, De Pauw G, Aertgeerts S, Valkenburg F, De Deyn PP, Verhaeghe J, Wyffels L, Van der Linden A, Staelens S, Verhoye M, Dedeurwaerdere S. Neuroimaging of Subacute Brain Inflammation and Microstructural Changes Predicts Long-Term Functional Outcome after Experimental Traumatic Brain Injury. J Neurotrauma 2018; 36:768-788. [PMID: 30032713 DOI: 10.1089/neu.2018.5704] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
There is currently a lack of prognostic biomarkers to predict the different sequelae following traumatic brain injury (TBI). The present study investigated the hypothesis that subacute neuroinflammation and microstructural changes correlate with chronic TBI deficits. Rats were subjected to controlled cortical impact (CCI) injury, sham surgery, or skin incision (naïve). CCI-injured (n = 18) and sham-operated rats (n = 6) underwent positron emission tomography (PET) imaging with the translocator protein 18 kDa (TSPO) radioligand [18F]PBR111 and diffusion tensor imaging (DTI) in the subacute phase (≤3 weeks post-injury) to quantify inflammation and microstructural alterations. CCI-injured, sham-operated, and naïve rats (n = 8) underwent behavioral testing in the chronic phase (5.5-10 months post-injury): open field and sucrose preference tests, two one-week video-electroencephalogram (vEEG) monitoring periods, pentylenetetrazole (PTZ) seizure susceptibility tests, and a Morris water maze (MWM) test. In vivo imaging revealed pronounced neuroinflammation, decreased fractional anisotropy, and increased diffusivity in perilesional cortex and ipsilesional hippocampus of CCI-injured rats. Behavioral analysis revealed disinhibition, anhedonia, increased seizure susceptibility, and impaired learning in CCI-injured rats. Subacute TSPO expression and changes in DTI metrics significantly correlated with several chronic deficits (Pearson's |r| = 0.50-0.90). Certain specific PET and DTI parameters had good sensitivity and specificity (area under the receiver operator characteristic [ROC] curve = 0.85-1.00) to distinguish between TBI animals with and without particular behavioral deficits. Depending on the investigated behavioral deficit, PET or DTI data alone, or the combination, could very well predict the variability in functional outcome data (adjusted R2 = 0.54-1.00). Taken together, both TSPO PET and DTI seem promising prognostic biomarkers to predict different chronic TBI sequelae.
Collapse
Affiliation(s)
- Stephan Missault
- 1 Experimental Laboratory of Translational Neuroscience and Otolaryngology, Faculty of Medicine and Health Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium .,2 Bio-Imaging Lab, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Cynthia Anckaerts
- 2 Bio-Imaging Lab, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Ines Blockx
- 2 Bio-Imaging Lab, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Steven Deleye
- 3 Molecular Imaging Center Antwerp, Faculty of Medicine and Health Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Debby Van Dam
- 4 Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Wilrijk, Belgium; Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen (UMCG) , Groningen, The Netherlands
| | - Nora Barriche
- 1 Experimental Laboratory of Translational Neuroscience and Otolaryngology, Faculty of Medicine and Health Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Glenn De Pauw
- 1 Experimental Laboratory of Translational Neuroscience and Otolaryngology, Faculty of Medicine and Health Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Stephanie Aertgeerts
- 1 Experimental Laboratory of Translational Neuroscience and Otolaryngology, Faculty of Medicine and Health Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Femke Valkenburg
- 4 Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Wilrijk, Belgium; Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen (UMCG) , Groningen, The Netherlands
| | - Peter Paul De Deyn
- 4 Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, University of Antwerp, Wilrijk, Belgium; Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen (UMCG) , Groningen, The Netherlands
| | - Jeroen Verhaeghe
- 3 Molecular Imaging Center Antwerp, Faculty of Medicine and Health Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Leonie Wyffels
- 3 Molecular Imaging Center Antwerp, Faculty of Medicine and Health Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium .,5 Department of Nuclear Medicine, University Hospital Antwerp , Edegem, Belgium
| | - Annemie Van der Linden
- 2 Bio-Imaging Lab, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Steven Staelens
- 3 Molecular Imaging Center Antwerp, Faculty of Medicine and Health Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Marleen Verhoye
- 2 Bio-Imaging Lab, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| | - Stefanie Dedeurwaerdere
- 6 Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, University of Antwerp , Wilrijk, Belgium
| |
Collapse
|
117
|
Szczygielski J, Glameanu C, Müller A, Klotz M, Sippl C, Hubertus V, Schäfer KH, Mautes AE, Schwerdtfeger K, Oertel J. Changes in Posttraumatic Brain Edema in Craniectomy-Selective Brain Hypothermia Model Are Associated With Modulation of Aquaporin-4 Level. Front Neurol 2018; 9:799. [PMID: 30333785 PMCID: PMC6176780 DOI: 10.3389/fneur.2018.00799] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 09/04/2018] [Indexed: 12/19/2022] Open
Abstract
Both hypothermia and decompressive craniectomy have been considered as a treatment for traumatic brain injury. In previous experiments we established a murine model of decompressive craniectomy and we presented attenuated edema formation due to focal brain cooling. Since edema development is regulated via function of water channel proteins, our hypothesis was that the effects of decompressive craniectomy and of hypothermia are associated with a change in aquaporin-4 (AQP4) concentration. Male CD-1 mice were assigned into following groups (n = 5): sham, decompressive craniectomy, trauma, trauma followed by decompressive craniectomy and trauma + decompressive craniectomy followed by focal hypothermia. After 24 h, magnetic resonance imaging with volumetric evaluation of edema and contusion were performed, followed by ELISA analysis of AQP4 concentration in brain homogenates. Additional histopathological analysis of AQP4 immunoreactivity has been performed at more remote time point of 28d. Correlation analysis revealed a relationship between AQP4 level and both volume of edema (r2 = 0.45, p < 0.01, **) and contusion (r2 = 0.41, p < 0.01, **) 24 h after injury. Aggregated analysis of AQP4 level (mean ± SEM) presented increased AQP4 concentration in animals subjected to trauma and decompressive craniectomy (52.1 ± 5.2 pg/mL, p = 0.01; *), but not to trauma, decompressive craniectomy and hypothermia (45.3 ± 3.6 pg/mL, p > 0.05; ns) as compared with animals subjected to decompressive craniectomy only (32.8 ± 2.4 pg/mL). However, semiquantitative histopathological analysis at remote time point revealed no significant difference in AQP4 immunoreactivity across the experimental groups. This suggests that AQP4 is involved in early stages of brain edema formation after surgical decompression. The protective effect of selective brain cooling may be related to change in AQP4 response after decompressive craniectomy. The therapeutic potential of this interaction should be further explored.
Collapse
Affiliation(s)
- Jacek Szczygielski
- Department of Neurosurgery, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany.,Institute of Neuropathology, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany.,Faculty of Medicine, University of Rzeszów, Rzeszów, Poland
| | - Cosmin Glameanu
- Department of Neurosurgery, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Andreas Müller
- Department of Radiology, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Markus Klotz
- Working Group Enteric Nervous System (AGENS), University of Applied Sciences Kaiserslautern, Kaiserslautern, Germany
| | - Christoph Sippl
- Department of Neurosurgery, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Vanessa Hubertus
- Department of Neurosurgery, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany.,Department of Neurosurgery, Charité University Medicine, Berlin, Germany
| | - Karl-Herbert Schäfer
- Working Group Enteric Nervous System (AGENS), University of Applied Sciences Kaiserslautern, Kaiserslautern, Germany
| | - Angelika E Mautes
- Department of Neurosurgery, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Karsten Schwerdtfeger
- Department of Neurosurgery, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Joachim Oertel
- Department of Neurosurgery, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| |
Collapse
|
118
|
da Silva Fiorin F, do Espírito Santo CC, Santos ARS, Fighera MR, Royes LFF. Implication of surgical procedure in the induction of headache and generalized painful sensation in a fluid percussion injury model in rats. J Neurosci Methods 2018; 307:23-30. [DOI: 10.1016/j.jneumeth.2018.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 05/29/2018] [Accepted: 06/12/2018] [Indexed: 12/27/2022]
|
119
|
Brady RD, Casillas-Espinosa PM, Agoston DV, Bertram EH, Kamnaksh A, Semple BD, Shultz SR. Modelling traumatic brain injury and posttraumatic epilepsy in rodents. Neurobiol Dis 2018; 123:8-19. [PMID: 30121231 DOI: 10.1016/j.nbd.2018.08.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/25/2018] [Accepted: 08/13/2018] [Indexed: 12/14/2022] Open
Abstract
Posttraumatic epilepsy (PTE) is one of the most debilitating and understudied consequences of traumatic brain injury (TBI). It is challenging to study the effects, underlying pathophysiology, biomarkers, and treatment of TBI and PTE purely in human patients for a number of reasons. Rodent models can complement human PTE studies as they allow for the rigorous investigation into the causal relationship between TBI and PTE, the pathophysiological mechanisms of PTE, the validation and implementation of PTE biomarkers, and the assessment of PTE treatments, in a tightly controlled, time- and cost-efficient manner in experimental subjects known to be experiencing epileptogenic processes. This article will review several common rodent models of TBI and/or PTE, including their use in previous studies and discuss their relative strengths, limitations, and avenues for future research to advance our understanding and treatment of PTE.
Collapse
Affiliation(s)
- Rhys D Brady
- Departments of Neuroscience and Medicine, Central Clinical School, Monash University, VIC 3004, Australia; Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, VIC 3052, Australia.
| | - Pablo M Casillas-Espinosa
- Departments of Neuroscience and Medicine, Central Clinical School, Monash University, VIC 3004, Australia; Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, VIC 3052, Australia.
| | - Denes V Agoston
- Anatomy, Physiology & Genetics, Uniformed Services University, Bethesda, MD 20814, USA
| | - Edward H Bertram
- Department of Neurology, University of Virginia, P.O. Box 800394, Charlottesville, VA 22908-0394, USA
| | - Alaa Kamnaksh
- Anatomy, Physiology & Genetics, Uniformed Services University, Bethesda, MD 20814, USA
| | - Bridgette D Semple
- Departments of Neuroscience and Medicine, Central Clinical School, Monash University, VIC 3004, Australia; Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, VIC 3052, Australia
| | - Sandy R Shultz
- Departments of Neuroscience and Medicine, Central Clinical School, Monash University, VIC 3004, Australia; Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, VIC 3052, Australia
| |
Collapse
|
120
|
Strides Toward Better Understanding of Post-Traumatic Headache Pathophysiology Using Animal Models. Curr Pain Headache Rep 2018; 22:67. [PMID: 30073545 DOI: 10.1007/s11916-018-0720-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW In recent years, the awareness of the detrimental impact of concussion and mild traumatic brain injuries (mTBI) is becoming more apparent. Concussive head trauma results in a constellation of cognitive and somatic symptoms of which post-traumatic headache is the most common. Our understanding of post-traumatic headache is limited by the paucity of well validated, characterized, and clinically relevant animal models with strong predictive validity. In this review, we aim to summarize and discuss current animal models of concussion/mTBI and related data that start to shed light on the pathophysiology of post-traumatic headache. RECENT FINDINGS Each of the models will be discussed in terms of their face, construct, and predictive validity as well as overall translational relevance to concussion, mTBI, and post-traumatic headache. Significant contributions to the pathophysiology of PTH garnered from these models are discussed as well as potential contributors to the development of chronic post-traumatic headache. Although post-traumatic headache is one of the most common symptoms following mild head trauma, there remains a disconnect between the study of mild traumatic brain injury and headache in the pre-clinical literature. A greater understanding of the relationship between these phenomena is currently needed to provide more insight into the increasing frequency of this debilitating condition in both military and civilian populations.
Collapse
|
121
|
Flygt J, Ruscher K, Norberg A, Mir A, Gram H, Clausen F, Marklund N. Neutralization of Interleukin-1β following Diffuse Traumatic Brain Injury in the Mouse Attenuates the Loss of Mature Oligodendrocytes. J Neurotrauma 2018; 35:2837-2849. [PMID: 29690837 PMCID: PMC6247990 DOI: 10.1089/neu.2018.5660] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Traumatic brain injury (TBI) commonly results in injury to the components of the white matter tracts, causing post-injury cognitive deficits. The myelin-producing oligodendrocytes (OLs) are vulnerable to TBI, although may potentially be replaced by proliferating oligodendrocyte progenitor cells (OPCs). The cytokine interleukin-1β (IL-1β) is a key mediator of the complex inflammatory response, and when neutralized in experimental TBI, behavioral outcome was improved. To evaluate the role of IL-1β on oligodendrocyte cell death and OPC proliferation, 116 adult male mice subjected to sham injury or the central fluid percussion injury (cFPI) model of traumatic axonal injury, were analyzed at two, seven, and 14 days post-injury. At 30 min post-injury, mice were randomly administered an IL-1β neutralizing or a control antibody. OPC proliferation (5-ethynyl 2'- deoxyuridine (EdU)/Olig2 co-labeling) and mature oligodendrocyte cell loss was evaluated in injured white matter tracts. Microglia/macrophages immunohistochemistry and ramification using Sholl analysis were also evaluated. Neutralizing IL-1β resulted in attenuated cell death, indicated by cleaved caspase-3 expression, and attenuated loss of mature OLs from two to seven days post-injury in brain-injured animals. IL-1β neutralization also attenuated the early, two day post-injury increase of microglia/macrophage immunoreactivity and altered their ramification. The proliferation of OPCs in brain-injured animals was not altered, however. Our data suggest that IL-1β is involved in the TBI-induced loss of OLs and early microglia/macrophage activation, although not the OPC proliferation. Attenuated oligodendrocyte cell loss may contribute to the improved behavioral outcome observed by IL-1β neutralization in this mouse model of diffuse TBI.
Collapse
Affiliation(s)
- Johanna Flygt
- 1 Department of Neuroscience, Section of Neurosurgery, Uppsala University , Uppsala, Sweden
| | - Karsten Ruscher
- 2 Novartis Institutes of Biomedical Research , Basel, Switzerland
| | - Amanda Norberg
- 1 Department of Neuroscience, Section of Neurosurgery, Uppsala University , Uppsala, Sweden
| | - Anis Mir
- 3 Lund University, Skane University Hospital , Department of Clinical Sciences Lund, Neurosurgery, Lund, Sweden
| | - Hermann Gram
- 3 Lund University, Skane University Hospital , Department of Clinical Sciences Lund, Neurosurgery, Lund, Sweden
| | - Fredrik Clausen
- 1 Department of Neuroscience, Section of Neurosurgery, Uppsala University , Uppsala, Sweden
| | - Niklas Marklund
- 1 Department of Neuroscience, Section of Neurosurgery, Uppsala University , Uppsala, Sweden .,3 Lund University, Skane University Hospital , Department of Clinical Sciences Lund, Neurosurgery, Lund, Sweden
| |
Collapse
|
122
|
Falnikar A, Stratton J, Lin R, Andrews CE, Tyburski A, Trovillion VA, Gottschalk C, Ghosh B, Iacovitti L, Elliott MB, Lepore AC. Differential Response in Novel Stem Cell Niches of the Brain after Cervical Spinal Cord Injury and Traumatic Brain Injury. J Neurotrauma 2018; 35:2195-2207. [PMID: 29471717 DOI: 10.1089/neu.2017.5497] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Populations of neural stem cells (NSCs) reside in a number of defined niches in the adult central nervous system (CNS) where they continually give rise to mature cell types throughout life, including newly born neurons. In addition to the prototypical niches of the subventricular zone (SVZ) and subgranular zone (SGZ) of the hippocampal dentate gyrus, novel stem cell niches that are also neurogenic have recently been identified in multiple midline structures, including circumventricular organs (CVOs) of the brain. These resident NSCs serve as a homeostatic source of new neurons and glial cells under intact physiological conditions. Importantly, they may also have the potential for reparative processes in pathological states such as traumatic spinal cord injury (SCI) and traumatic brain injury (TBI). As the response in these novel CVO stem cell niches has been characterized after stroke but not following SCI or TBI, we quantitatively assessed cell proliferation and the neuronal and glial lineage fate of resident NSCs in three CVO nuclei-area postrema (AP), median eminence (ME), and subfornical organ (SFO) -in rat models of cervical contusion-type SCI and controlled cortical impact (CCI)-induced TBI. Using bromodeoxyuridine (BrdU) labeling of proliferating cells, we find that TBI significantly enhanced proliferation in AP, ME, and SFO, whereas cervical SCI had no effects at early or chronic time-points post-injury. In addition, SCI did not alter NSC differentiation profile into doublecortin-positive neuroblasts, GFAP-expressing astrocytes, or Olig2-labeled cells of the oligodendrocyte lineage within AP, ME, or SFO at both time-points. In contrast, CCI induced a pronounced increase in Sox2- and doublecortin-labeled cells in the AP and Iba1-labeled microglia in the SFO. Lastly, plasma derived from CCI animals significantly increased NSC expansion in an in vitro neurosphere assay, whereas plasma from SCI animals did not exert such an effect, suggesting that signaling factors present in blood may be relevant to stimulating CVO niches after CNS injury and may explain the differential in vivo effects of SCI and TBI on the novel stem cell niches.
Collapse
Affiliation(s)
- Aditi Falnikar
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Jarred Stratton
- 2 Department of Neurological Surgery, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Ruihe Lin
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Carrie E Andrews
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Ashley Tyburski
- 2 Department of Neurological Surgery, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Victoria A Trovillion
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Chelsea Gottschalk
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Biswarup Ghosh
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Lorraine Iacovitti
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Melanie B Elliott
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania.,2 Department of Neurological Surgery, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Angelo C Lepore
- 1 Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University , Philadelphia, Pennsylvania
| |
Collapse
|
123
|
Letson HL, Dobson GP. Adenosine, lidocaine, and Mg2+ (ALM) resuscitation fluid protects against experimental traumatic brain injury. J Trauma Acute Care Surg 2018; 84:908-916. [PMID: 29554045 DOI: 10.1097/ta.0000000000001874] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Currently, no drug therapy prevents secondary injury progression after traumatic brain injury (TBI). Our aim was to investigate the effects of small-volume intravenous adenosine, lidocaine, and Mg (ALM) resuscitation fluid after moderate TBI in a rat fluid-percussion injury model. METHODS Anesthetized, mechanically ventilated male Sprague-Dawley rats (449 ± 5 g) were randomly assigned to one of four groups: (1) sham (craniotomy without TBI), (2) no-treatment, (3) saline-control, or (4) ALM therapy groups (all n = 16). A subdural probe was implanted in eight animals per group to measure cerebral blood flow. Fifteen minutes after moderate TBI was induced with lateral fluid percussion injury (2.57 atm), a single 3% NaCl ± ALM bolus (0.7 mL/kg) was injected intravenously, and after 60 minutes (Phase 1), 0.9% NaCl ± ALM stabilization "drip" (0.5 mL/kg per hour) was administered for 3 hours (Phase 2). RESULTS Mortality (without subdural brain probe) was 25% (saline controls) and 0% (ALM). Sixty minutes after bolus, ALM significantly increased cardiac function, cortical blood flow (CBF; approximately threefold) and blunted systemic inflammation compared to saline controls. Three hours after infusion drip, ALM improved left ventricular function, supported higher CBF, decreased proinflammatory cytokines systemically (IL-1β, tumor necrosis factor α, and regulated on activation, normal T cell expressed and secreted [RANTES]), increased anti-inflammatory cytokines in brain tissue (IL-10, IL-4), lowered brain injury markers (neuron-specific enolase, Syndecan-1, HMGB-1), reduced coagulopathy, increased platelet aggregation, and maintained baseline fibrinogen levels. Saline-controls were proinflammatory (brain, heart, lung, and blood) and hypocoagulable with neurogenic enlargement of the right side of the heart. Survival time significantly correlated with plasma neuron-specific enolase (p = 0.001) and CBF at 180 minutes (p = 0.009), and CBF correlated with brain anti-inflammatory cytokines (p = 0.001-0.034). CONCLUSION After moderate TBI, ALM resuscitation fluid increased survival and protected against early secondary injury by reducing coagulopathy, inflammation, and platelet dysfunction.
Collapse
Affiliation(s)
- Hayley L Letson
- From the Heart, Trauma and Sepsis Research Laboratory (H.LL, G.P.D.), College of Medicine and Dentistry. James Cook University, Townsville, Queensland, Australia
| | | |
Collapse
|
124
|
Shoffstall AJ, Paiz J, Miller D, Rial G, Willis M, Menendez D, Hostler S, Capadona JR. Potential for thermal damage to the blood-brain barrier during craniotomy: implications for intracortical recording microelectrodes. J Neural Eng 2018; 15:034001. [PMID: 29205169 PMCID: PMC6482047 DOI: 10.1088/1741-2552/aa9f32] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Our objective was to determine how readily disruption of the blood-brain barrier (BBB) occurred as a result of bone drilling during a craniotomy to implant microelectrodes in rat cortex. While the phenomenon of heat production during bone drilling is well known, practices to evade damage to the underlying brain tissue are inconsistently practiced and reported in the literature. APPROACH We conducted a review of the intracortical microelectrode literature to summarize typical approaches to mitigate drill heating during rodent craniotomies. Post mortem skull-surface and transient brain-surface temperatures were experimentally recorded using an infrared camera and thermocouple, respectively. A number of drilling conditions were tested, including varying drill speed and continuous versus intermittent contact. In vivo BBB permeability was assayed 1 h after the craniotomy procedure using Evans blue dye. MAIN RESULTS Of the reviewed papers that mentioned methods to mitigate thermal damage during craniotomy, saline irrigation was the most frequently cited (in six of seven papers). In post mortem tissues, we observed increases in skull-surface temperature ranging from +3 °C to +21 °C, dependent on drill speed. In vivo, pulsed-drilling (2 s-on/2 s-off) and slow-drilling speeds (1000 r.p.m.) were the most effective methods we studied to mitigate heating effects from drilling, while inconclusive results were obtained with saline irrigation. SIGNIFICANCE Neuroinflammation, initiated by damage to the BBB and perpetuated by the foreign body response, is thought to play a key role in premature failure of intracortical recording microelectrodes. This study demonstrates the extreme sensitivity of the BBB to overheating caused by bone drilling. To avoid damage to the BBB, the authors recommend that craniotomies be drilled with slow speeds and/or with intermittent drilling with complete removal of the drill from the skull during 'off' periods. While saline alone was ineffective at preventing overheating, its use is still recommended to remove bone dust from the surgical site and to augment other cooling methods.
Collapse
Affiliation(s)
- Andrew J. Shoffstall
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44016
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, 10701 East Blvd, 151 W/APT, Cleveland, OH 44106-1702, USA
| | - Jen Paiz
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44016
| | - David Miller
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44016
| | - Griffin Rial
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44016
| | - Mitchell Willis
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44016
| | - Dhariyat Menendez
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44016
| | - Stephen Hostler
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, OH 44106
| | - Jeffrey R. Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44016
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, 10701 East Blvd, 151 W/APT, Cleveland, OH 44106-1702, USA
| |
Collapse
|
125
|
Hylin MJ, Holden RC, Smith AC, Logsdon AF, Qaiser R, Lucke-Wold BP. Juvenile Traumatic Brain Injury Results in Cognitive Deficits Associated with Impaired Endoplasmic Reticulum Stress and Early Tauopathy. Dev Neurosci 2018; 40:175-188. [PMID: 29788004 PMCID: PMC6376969 DOI: 10.1159/000488343] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 03/12/2018] [Indexed: 02/05/2023] Open
Abstract
The leading cause of death in the juvenile population is trauma, and in particular neurotrauma. The juvenile brain response to neurotrauma is not completely understood. Endoplasmic reticulum (ER) stress has been shown to contribute to injury expansion and behavioral deficits in adult rodents and furthermore has been seen in adult postmortem human brains diagnosed with chronic traumatic encephalopathy. Whether endoplasmic reticulum stress is increased in juveniles with traumatic brain injury (TBI) is poorly delineated. We investigated this important topic using a juvenile rat controlled cortical impact (CCI) model. We proposed that ER stress would be significantly increased in juvenile rats following TBI and that this would correlate with behavioral deficits using a juvenile rat model. A juvenile rat (postnatal day 28) CCI model was used. Binding immunoglobulin protein (BiP) and C/EBP homologous protein (CHOP) were measured at 4 h in the ipsilateral pericontusion cortex. Hypoxia-inducible factor (HIF)-1α was measured at 48 h and tau kinase measured at 1 week and 30 days. At 4 h following injury, BiP and CHOP (markers of ER stress) were significantly elevated in rats exposed to TBI. We also found that HIF-1α was significantly upregulated 48 h following TBI showing delayed hypoxia. The early ER stress activation was additionally asso-ciated with the activation of a known tau kinase, glycogen synthase kinase-3β (GSK-3β), by 1 week. Tau oligomers measured by R23 were significantly increased by 30 days following TBI. The biochemical changes following TBI were associated with increased impulsive-like or anti-anxiety behavior measured with the elevated plus maze, deficits in short-term memory measured with novel object recognition, and deficits in spatial memory measured with the Morris water maze in juvenile rats exposed to TBI. These results show that ER stress was increased early in juvenile rats exposed to TBI, that these rats developed tau oligomers over the course of 30 days, and that they had significant short-term and spatial memory deficits following injury.
Collapse
Affiliation(s)
- Michael J. Hylin
- Neurotrauma and Rehabilitation Laboratory, Department of Psychology, Southern Illinois University, Carbondale, IL, USA
| | - Ryan C. Holden
- Neurotrauma and Rehabilitation Laboratory, Department of Psychology, Southern Illinois University, Carbondale, IL, USA
| | - Aidan C. Smith
- Neurotrauma and Rehabilitation Laboratory, Department of Psychology, Southern Illinois University, Carbondale, IL, USA
| | - Aric F. Logsdon
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Rabia Qaiser
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Brandon P. Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
| |
Collapse
|
126
|
Abstract
Mild traumatic brain injury (mTBI) represents a significant public healthcare concern, accounting for the majority of all head injuries. While symptoms are generally transient, some patients go on to experience long-term cognitive impairments and additional mild impacts can result in exacerbated and persisting negative outcomes. To date, studies using a range of experimental models have reported chronic behavioral deficits in the presence of axonal injury and inflammation following repeated mTBI; assessments of oxidative stress and myelin pathology have thus far been limited. However, some models employed induced acute focal damage more suggestive of moderate–severe brain injury and are therefore not relevant to repeated mTBI. Given that the nature of mechanical loading in TBI is implicated in downstream pathophysiological changes, the mechanisms of damage and chronic consequences of single and repeated closed-head mTBI remain to be fully elucidated. This review covers literature on potential mechanisms of damage following repeated mTBI, integrating known mechanisms of pathology underlying moderate–severe TBIs, with recent studies on adult rodent models relevant to direct impact injuries rather than blast-induced damage. Pathology associated with excitotoxicity and cerebral blood flow-metabolism uncoupling, oxidative stress, cell death, blood-brain barrier dysfunction, astrocyte reactivity, microglial activation, diffuse axonal injury, and dysmyelination is discussed, followed by a summary of functional deficits and preclinical assessments of therapeutic strategies. Comprehensive characterization of the pathology underlying delayed and persisting deficits following repeated mTBI is likely to facilitate further development of therapeutic strategies to limit long-term sequelae.
Collapse
Affiliation(s)
- Brooke Fehily
- 1 Experimental and Regenerative Neurosciences, School of Biological sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Melinda Fitzgerald
- 1 Experimental and Regenerative Neurosciences, School of Biological sciences, The University of Western Australia, Perth, Western Australia, Australia.,2 Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia.,3 Perron Institute for Neurological and Translational Science, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
| |
Collapse
|
127
|
Bloch K, Gil-Ad I, Vanichkin A, Hornfeld SH, Koroukhov N, Taler M, Vardi P, Weizman A. Intracerebroventricular Streptozotocin Induces Obesity and Dementia in Lewis Rats. J Alzheimers Dis 2018; 60:121-136. [PMID: 28800326 DOI: 10.3233/jad-161289] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Animal models of dementia associated with metabolic abnormalities play an important role in understanding the bidirectional relationships between these pathologies. Rodent strains develop cognitive dysfunctions without alteration of peripheral metabolism following intracerebroventricular administration of streptozotocin (icv-STZ). OBJECTIVE We aimed to estimate the effect of icv-STZ on cognitive functions and peripheral metabolism in Lewis rats, which are rarely used for the induction of cognitive abnormalities. METHODS Inbred adult Lewis rats were treated with single icv-STZ (3 mg/kg). Cognitive functions were assessed using Morris water maze (MWM) test and locomotion by the Open Field test. Metabolic alterations were studied using histological and biochemical analysis of brain and peripheral tissues. RESULTS The icv-STZ induced rapid weight decline during the first two weeks. Thereafter, the rats showed an accelerated weight gain. Three months after the icv-STZ treatment, the rats were severely obese and revealed fatty liver, pancreatic islet hypertrophy, significantly elevated levels of blood insulin, leptin, and adiponectin, but intact peripheral glucose homeostasis. The icv-STZ rats expressed amyloid-β deposits in blood vessels of leptomeningeal area, microgliosis, astrogliosis, and spongiosis in fimbria-fornix area of hippocampus. Locomotor activities of icv-STZ treated and sham-operated rats were similar. In the MWM test, the icv-STZ treated rats demonstrated severely impaired spatial learning during both acquisition and reversal phases. CONCLUSIONS Icv-STZ treated Lewis rats develop severe dementia associated with obesity and peripheral metabolic abnormalities. This animal model may be useful for exploring the pathophysiological relationship between obesity and dementia and provides a new tool for development of effective therapy.
Collapse
Affiliation(s)
- Konstantin Bloch
- Laboratory of Diabetes and Obesity Research, Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petah Tikva, Israel
| | - Irit Gil-Ad
- Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petah Tikva, Israel
| | - Alexey Vanichkin
- Laboratory of Transplantation, Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petah Tikva, Israel
| | - Shay Henry Hornfeld
- Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petah Tikva, Israel
| | - Nickolay Koroukhov
- Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petah Tikva, Israel
| | - Michal Taler
- Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petah Tikva, Israel
| | - Pnina Vardi
- Laboratory of Diabetes and Obesity Research, Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petah Tikva, Israel
| | - Abraham Weizman
- Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petah Tikva, Israel.,Research Unit, Geha Mental Health Center, Petah Tikva, Israel
| |
Collapse
|
128
|
Nichols J, Bjorklund GR, Newbern J, Anderson T. Parvalbumin fast-spiking interneurons are selectively altered by paediatric traumatic brain injury. J Physiol 2018; 596:1277-1293. [PMID: 29333742 PMCID: PMC5878227 DOI: 10.1113/jp275393] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 12/19/2017] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Traumatic brain injury (TBI) in children remains a leading cause of death and disability and it remains poorly understood why children have worse outcomes and longer recover times. TBI has shown to alter cortical excitability and inhibitory drive onto excitatory neurons, yet few studies have directly examined changes to cortical interneurons. This is addressed in the present study using a clinically relevant model of severe TBI (controlled cortical impact) in interneuron cell type specific Cre-dependent mice. Mice subjected to controlled cortical impact exhibit specific loss of parvalbumin (PV) but not somatostatin immunoreactivity and cell density in the peri-injury zone. PV interneurons are primarily of a fast-spiking (FS) phenotype that persisted in the peri-injury zone but received less frequent inhibitory and stronger excitatory post-synaptic currents. The targeted loss of PV-FS interneurons appears to be distinct from previous reports in adult mice suggesting that TBI-induced pathophysiology is dependent on the age at time of impact. ABSTRACT Paediatric traumatic brain injury (TBI) is a leading cause of death and disability in children. Traditionally, ongoing neurodevelopment and neuroplasticity have been considered to confer children with an advantage following TBI. However, recent findings indicate that the paediatric brain may be more sensitive to brain injury. Inhibitory interneurons are essential for proper cortical function and are implicated in the pathophysiology of TBI, yet few studies have directly investigated TBI-induced changes to interneurons themselves. Accordingly, in the present study, we examine how inhibitory neurons are altered following controlled cortical impact (CCI) in juvenile mice with targeted Cre-dependent fluorescence labelling of interneurons (Vgat:Cre/Ai9 and PV:Cre/Ai6). Although CCI failed to alter the number of excitatory neurons or somatostatin-expressing interneurons in the peri-injury zone, it significantly decreased the density of parvalbumin (PV) immunoreactive cells by 71%. However, PV:Cre/Ai6 mice subjected to CCI showed a lower extent of fluorescence labelled cell loss. PV interneurons are predominantly of a fast-spiking (FS) phenotype and, when recorded electrophysiologically from the peri-injury zone, exhibited intrinsic properties similar to those of control neurons. Synaptically, CCI induced a decrease in inhibitory drive onto FS interneurons combined with an increase in the strength of excitatory events. The results of the present study indicate that CCI induced both a loss of PV interneurons and an even greater loss of PV expression. This suggests caution is required when interpreting changes in PV immunoreactivity alone as direct evidence of interneuronal loss. Furthermore, in contrast to reports in adults, TBI in the paediatric brain selectively alters PV-FS interneurons, primarily resulting in a loss of interneuronal inhibition.
Collapse
Affiliation(s)
- Joshua Nichols
- University of ArizonaCollege of Medicine – PhoenixPhoenixAZUSA
- School of Life SciencesArizona State UniversityAZUSA
| | | | - Jason Newbern
- School of Life SciencesArizona State UniversityAZUSA
| | - Trent Anderson
- University of ArizonaCollege of Medicine – PhoenixPhoenixAZUSA
| |
Collapse
|
129
|
Bedi SS, Aertker BM, Liao GP, Caplan HW, Bhattarai D, Mandy F, Mandy F, Fernandez LG, Zelnick P, Mitchell MB, Schiffer W, Johnson M, Denson E, Prabhakara K, Xue H, Smith P, Uray K, Olson SD, Mays RW, Cox CS. Therapeutic time window of multipotent adult progenitor therapy after traumatic brain injury. J Neuroinflammation 2018; 15:84. [PMID: 29548333 PMCID: PMC5856201 DOI: 10.1186/s12974-018-1122-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 03/08/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a major cause of death and disability. TBI results in a prolonged secondary central neuro-inflammatory response. Previously, we have demonstrated that multiple doses (2 and 24 h after TBI) of multipotent adult progenitor cells (MAPC) delivered intravenously preserve the blood-brain barrier (BBB), improve spatial learning, and decrease activated microglia/macrophages in the dentate gyrus of the hippocampus. In order to determine if there is an optimum treatment window to preserve the BBB, improve cognitive behavior, and attenuate the activated microglia/macrophages, we administered MAPC at various clinically relevant intervals. METHODS We administered two injections intravenously of MAPC treatment at hours 2 and 24 (2/24), 6 and 24 (6/24), 12 and 36 (12/36), or 36 and 72 (36/72) post cortical contusion injury (CCI) at a concentration of 10 million/kg. For BBB experiments, animals that received MAPC at 2/24, 6/24, and 12/36 were euthanized 72 h post injury. The 36/72 treated group was harvested at 96 h post injury. RESULTS Administration of MAPC resulted in a significant decrease in BBB permeability when administered at 2/24 h after TBI only. For behavior experiments, animals were harvested post behavior paradigm. There was a significant improvement in spatial learning (120 days post injury) when compared to cortical contusion injury (CCI) in groups when MAPC was administered at or before 24 h. In addition, there was a significant decrease in activated microglia/macrophages in the dentate gyrus of hippocampus of the treated group (2/24) only when compared to CCI. CONCLUSIONS Intravenous injections of MAPC at or before 24 h after CCI resulted in improvement of the BBB, improved cognitive behavior, and attenuated activated microglia/macrophages in the dentate gyrus.
Collapse
Affiliation(s)
- Supinder S Bedi
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA.
| | - Benjamin M Aertker
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - George P Liao
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Henry W Caplan
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Deepa Bhattarai
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Fanni Mandy
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Franciska Mandy
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Luis G Fernandez
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Pamela Zelnick
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Matthew B Mitchell
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Walter Schiffer
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Margaret Johnson
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Emma Denson
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Karthik Prabhakara
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Hasen Xue
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Philippa Smith
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Karen Uray
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | - Scott D Olson
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA
| | | | - Charles S Cox
- Departments of Pediatric Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA.,Departments of Surgery, University of Texas, Health Science Center at Houston, Houston, TX, USA.,Michael E DeBakey Institute for Comparative Cardiovascular Science and Biomedical Devices and Athersys, Inc., Cleveland, OH, USA
| |
Collapse
|
130
|
Garg C, Seo JH, Ramachandran J, Loh JM, Calderon F, Contreras JE. Trovafloxacin attenuates neuroinflammation and improves outcome after traumatic brain injury in mice. J Neuroinflammation 2018; 15:42. [PMID: 29439712 PMCID: PMC5812039 DOI: 10.1186/s12974-018-1069-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 01/17/2018] [Indexed: 12/20/2022] Open
Abstract
Background Trovafloxacin is a broad-spectrum antibiotic, recently identified as an inhibitor of pannexin-1 (Panx1) channels. Panx1 channels are important conduits for the adenosine triphosphate (ATP) release from live and dying cells that enhances the inflammatory response of immune cells. Elevated extracellular levels ATP released upon injury activate purinergic pathways in inflammatory cells that promote migration, proliferation, phagocytosis, and apoptotic signals. Here, we tested whether trovafloxacin administration attenuates the neuroinflammatory response and improves outcomes after brain trauma. Methods The murine controlled cortical impact (CCI) model was used to determine whether in vivo delivery of trovafloxacin has anti-inflammatory and neuroprotective actions after brain trauma. Locomotor deficit was assessed using the rotarod test. Levels of tissue damage markers and inflammation were measured using western blot, qPCR, and immunofluorescence. In vitro assays were used to evaluate whether trovafloxacin blocks ATP release and cell migration in a chemotactic-stimulated microglia cell line. Results Trovafloxacin treatment of CCI-injured mice significantly reduced tissue damage markers and improved locomotor deficits. In addition, trovafloxacin treatment significantly reduced mRNA levels of several pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α), which correlates with an overall reduction in the accumulation of inflammatory cell types (neutrophils, microglia/macrophages, and astroglia) at the injury zone. To determine whether trovafloxacin exerted these effects by direct action on immune cells, we evaluated its effect on ATP release and cell migration using a chemotactic-stimulated microglial cell line. We found that trovafloxacin significantly inhibited both ATP release and migration of these cells. Conclusion Our results show that trovafloxacin administration has pronounced anti-inflammatory and neuroprotective effects following brain injury. These findings lay the foundation for future studies to directly test a role for Panx1 channels in pathological inflammation following brain trauma. Electronic supplementary material The online version of this article (10.1186/s12974-018-1069-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Charu Garg
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Joon Ho Seo
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Jayalakshmi Ramachandran
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Ji Meng Loh
- Department of Mathematical Sciences, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
| | - Frances Calderon
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, 185 South Orange Ave, Newark, NJ, 07103, USA.
| | - Jorge E Contreras
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, 185 South Orange Ave, Newark, NJ, 07103, USA.
| |
Collapse
|
131
|
Sangobowale MA, Grin'kina NM, Whitney K, Nikulina E, St Laurent-Ariot K, Ho JS, Bayzan N, Bergold PJ. Minocycline plus N-Acetylcysteine Reduce Behavioral Deficits and Improve Histology with a Clinically Useful Time Window. J Neurotrauma 2018; 35:907-917. [PMID: 29187031 DOI: 10.1089/neu.2017.5348] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
There are no drugs to manage traumatic brain injury (TBI) presently. A major problem in developing therapeutics is that drugs to manage TBI lack sufficient potency when dosed within a clinically relevant time window. Previous studies have shown that minocycline (MINO, 45 mg/kg) plus N-acetylcysteine (NAC, 150 mg/kg) synergistically improved cognition and memory, modulated inflammation, and prevented loss of oligodendrocytes that remyelinated damaged white matter when first dosed 1 h after controlled cortical impact (CCI) in rats. We show that MINO (45 mg/kg) plus NAC (150 mg/kg) also prevent brain injury in a mouse closed head injury (CHI) TBI model. Using the CHI model, the concentrations of MINO and NAC were titrated to determine that MINO (22.5 mg/kg) plus NAC (75 mg/kg) was more potent than the original formulation. MINO (22.5 mg/kg) plus NAC (75 mg/kg) also limited injury in the rat CCI model. The therapeutic time window of MINO plus NAC was then tested in the CHI and CCI models. Mice and rats could acquire an active place avoidance task when MINO plus NAC was first dosed at 12 h post-injury. A first dose at 12 h also limited gray matter injury in the hippocampus and preserved myelin in multiple white matter tracts. Mice and rats acquired Barnes maze when MINO plus NAC was first dosed at 24 h post-injury. These data suggest that MINO (22.5 mg/kg) plus NAC (75 mg/kg) remain potent when dosed at clinically useful time windows. Both MINO and NAC are drugs approved by the Food and Drug Administration and have been administered safely to patients in clinical trials at the doses in the new formulation. This suggests that the drug combination of MINO plus NAC may be effective in treating patients with TBI.
Collapse
Affiliation(s)
| | - Natalia M Grin'kina
- Department of Physiology and Pharmacology, SUNY-Downstate Medical Center, Brooklyn, New York
| | - Kristen Whitney
- School of Graduate Studies, SUNY-Downstate Medical Center, Brooklyn, New York
| | - Elena Nikulina
- Department of Physiology and Pharmacology, SUNY-Downstate Medical Center, Brooklyn, New York
| | - Karrah St Laurent-Ariot
- Department of Physiology and Pharmacology, SUNY-Downstate Medical Center, Brooklyn, New York
| | - Johnson S Ho
- Department of Physiology and Pharmacology, SUNY-Downstate Medical Center, Brooklyn, New York
| | - Narek Bayzan
- Department of Physiology and Pharmacology, SUNY-Downstate Medical Center, Brooklyn, New York
| | - Peter J Bergold
- Department of Physiology and Pharmacology, SUNY-Downstate Medical Center, Brooklyn, New York.,Robert F. Furchgott Center for Neural and Behavioral Science, SUNY-Downstate Medical Center, Brooklyn, New York
| |
Collapse
|
132
|
Salehi A, Jullienne A, Baghchechi M, Hamer M, Walsworth M, Donovan V, Tang J, Zhang JH, Pearce WJ, Obenaus A. Up-regulation of Wnt/β-catenin expression is accompanied with vascular repair after traumatic brain injury. J Cereb Blood Flow Metab 2018; 38:274-289. [PMID: 29160735 PMCID: PMC5951019 DOI: 10.1177/0271678x17744124] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Recent data suggest that repairing the cerebral vasculature after traumatic brain injury (TBI) may help to improve functional recovery. The Wnt/β-catenin signaling pathway promotes blood vessel formation during vascular development, but its role in vascular repair after TBI remains elusive. In this study, we examined how the cerebral vasculature responds to TBI and the role of Wnt/β-catenin signaling in vascular repair. We induced a moderate controlled cortical impact in adult mice and performed vessel painting to visualize the vascular alterations in the brain. Brain tissue around the injury site was assessed for β-catenin and vascular markers. A Wnt transgenic mouse line was utilized to evaluate Wnt gene expression. We report that TBI results in vascular loss followed by increases in vascular structure at seven days post injury (dpi). Immature, non-perfusing vessels were evident in the tissue around the injury site. β-catenin protein expression was significantly reduced in the injury site at 7 dpi. However, there was an increase in β-catenin expression in perilesional vessels at 1 and 7 dpi. Similarly, we found increased number of Wnt-GFP-positive vessels after TBI. Our findings suggest that Wnt/β-catenin expression contributes to the vascular repair process after TBI.
Collapse
Affiliation(s)
- Arjang Salehi
- 1 Cell, Molecular and Developmental Biology Program, 8790 University of California, Riverside , CA, USA.,2 Department of Pediatrics, 4608 Loma Linda University , Loma Linda, CA, USA
| | - Amandine Jullienne
- 2 Department of Pediatrics, 4608 Loma Linda University , Loma Linda, CA, USA
| | - Mohsen Baghchechi
- 2 Department of Pediatrics, 4608 Loma Linda University , Loma Linda, CA, USA
| | - Mary Hamer
- 2 Department of Pediatrics, 4608 Loma Linda University , Loma Linda, CA, USA
| | - Mark Walsworth
- 2 Department of Pediatrics, 4608 Loma Linda University , Loma Linda, CA, USA
| | - Virginia Donovan
- 2 Department of Pediatrics, 4608 Loma Linda University , Loma Linda, CA, USA
| | - Jiping Tang
- 4 Department of Physiology and Pharmacology, School of Medicine, 4608 Loma Linda University , Loma Linda, CA, USA
| | - John H Zhang
- 4 Department of Physiology and Pharmacology, School of Medicine, 4608 Loma Linda University , Loma Linda, CA, USA.,5 Department of Anesthesiology, School of Medicine, 4608 Loma Linda University , Loma Linda, CA, USA.,6 Department of Neurosurgery, School of Medicine, 4608 Loma Linda University , Loma Linda, CA, USA
| | - William J Pearce
- 4 Department of Physiology and Pharmacology, School of Medicine, 4608 Loma Linda University , Loma Linda, CA, USA.,7 Center for Perinatal Biology, 4608 Loma Linda University , Loma Linda, CA, USA
| | - Andre Obenaus
- 1 Cell, Molecular and Developmental Biology Program, 8790 University of California, Riverside , CA, USA.,2 Department of Pediatrics, 4608 Loma Linda University , Loma Linda, CA, USA.,3 Department of Pediatrics, 12219 University of California, Irvine , CA, USA
| |
Collapse
|
133
|
Flygt J, Clausen F, Marklund N. Diffuse traumatic brain injury in the mouse induces a transient proliferation of oligodendrocyte progenitor cells in injured white matter tracts. Restor Neurol Neurosci 2018; 35:251-263. [PMID: 27768001 DOI: 10.3233/rnn-160675] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Injury to the white matter may lead to impaired neuronal signaling and is commonly observed following traumatic brain injury (TBI). Although endogenous repair of TBI-induced white matter pathology is limited, oligodendrocyte progenitor cells (OPCs) may be stimulated to proliferate and regenerate functionally myelinating oligodendrocytes. Even though OPCs are present throughout the adult brain, little is known about their proliferative activity following axonal injury caused by TBI. OBJECTIVE We hypothesized that central fluid percussion injury (cFPI) in mice, a TBI model causing wide-spread axonal injury, results in OPC proliferation. METHODS Proliferation of OPCs was evaluated in 27 cFPI mice using 5-ethynyl-2'-deoxyuridine (EdU) labeling and a cell proliferation assay at 2 (n = 9), 7 (n = 8) and 21 (n = 10) days post injury (dpi). Sham-injured mice (n = 14) were used as controls. OPC proliferation was quantified by immunohistochemistry using the OPC markers NG2 and Olig2 in several white matter loci including the corpus callosum, external capsule, fimbriae, the internal capsule and cerebral peduncle. RESULTS The number of EdU/DAPI/Olig2-positive cells were increased in the cFPI group compared to sham-injured animals at 7 days post-injury (dpi; p≤0.05) in the majority of white matter regions. The OPC proliferation had subsided by 21 dpi. The number of EdU/DAPI/NG2 cells was also increase at 7 dpi in the external capsule and fimbriae. CONCLUSION These results suggest that traumatic axonal injury in the mouse induces a transient proliferative response of residing OPCs. These proliferating OPCs may replace dead oligodendrocytes and contribute to remyelination, which needs evaluation in future studies.
Collapse
|
134
|
Krämer T, Grob T, Menzel L, Hirnet T, Griemert E, Radyushkin K, Thal SC, Methner A, Schaefer MKE. Dimethyl fumarate treatment after traumatic brain injury prevents depletion of antioxidative brain glutathione and confers neuroprotection. J Neurochem 2017; 143:523-533. [DOI: 10.1111/jnc.14220] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/04/2017] [Accepted: 09/08/2017] [Indexed: 12/26/2022]
Affiliation(s)
- Tobias Krämer
- Department of Anesthesiology; University Medical Center of the Johannes Gutenberg-University Mainz; Mainz Germany
| | - Theresa Grob
- Department of Anesthesiology; University Medical Center of the Johannes Gutenberg-University Mainz; Mainz Germany
| | - Lutz Menzel
- Department of Anesthesiology; University Medical Center of the Johannes Gutenberg-University Mainz; Mainz Germany
| | - Tobias Hirnet
- Department of Anesthesiology; University Medical Center of the Johannes Gutenberg-University Mainz; Mainz Germany
| | - Eva Griemert
- Department of Anesthesiology; University Medical Center of the Johannes Gutenberg-University Mainz; Mainz Germany
| | - Konstantin Radyushkin
- Mouse Behavior Unit; Focus Program Translational Neurosciences (FTN); Johannes Gutenberg-University of Mainz; Mainz Germany
- Focus Program Translational Neurosciences (FTN); Johannes Gutenberg-University of Mainz; Mainz Germany
| | - Serge C. Thal
- Department of Anesthesiology; University Medical Center of the Johannes Gutenberg-University Mainz; Mainz Germany
- Focus Program Translational Neurosciences (FTN); Johannes Gutenberg-University of Mainz; Mainz Germany
| | - Axel Methner
- Focus Program Translational Neurosciences (FTN); Johannes Gutenberg-University of Mainz; Mainz Germany
- Department of Neurology; University Medical Center of the Johannes Gutenberg-University Mainz; Mainz Germany
| | - Michael K. E. Schaefer
- Department of Anesthesiology; University Medical Center of the Johannes Gutenberg-University Mainz; Mainz Germany
- Focus Program Translational Neurosciences (FTN); Johannes Gutenberg-University of Mainz; Mainz Germany
| |
Collapse
|
135
|
Dean DD, Frank JA, Turtzo LC. Controlled Cortical Impact in the Rat. ACTA ACUST UNITED AC 2017; 81:9.62.1-9.62.12. [PMID: 29058772 DOI: 10.1002/cpns.37] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability world-wide. Following initial injury, TBI patients can face long-term disability in the form of cognitive, physical, and psychological deficits, depending on the severity and location of injury. This results in an economic burden in the United States estimated to be $60 billion due to health-care costs and loss of productivity. TBI is a significant area of active research interest for both military and civilian medicine. Numerous pre-clinical animal models of TBI are used to characterize the anatomical and physiological pathways involved and to evaluate therapeutic interventions. Due to its flexibility and scalability, controlled cortical impact (CCI) is one of the most commonly used preclinical TBI models. This unit provides a basic CCI protocol performed in the rat. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Dana D Dean
- Neuroscience Institute, Baylor Scott & White Health, Temple, Texas
| | - Joseph A Frank
- Frank Laboratory, National Institutes of Health, Clinical Center and National Institutes of Biomedical Imaging and Bioengineering, Bethesda, Maryland
| | - L Christine Turtzo
- National Institute of Neurological Disease and Stroke, Bethesda, Maryland
| |
Collapse
|
136
|
Anthonymuthu TS, Kenny EM, Amoscato AA, Lewis J, Kochanek PM, Kagan VE, Bayır H. Global assessment of oxidized free fatty acids in brain reveals an enzymatic predominance to oxidative signaling after trauma. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2601-2613. [PMID: 28347845 PMCID: PMC5612836 DOI: 10.1016/j.bbadis.2017.03.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/15/2017] [Accepted: 03/23/2017] [Indexed: 12/31/2022]
Abstract
Traumatic brain injury (TBI) is a major health problem associated with significant morbidity and mortality. The pathophysiology of TBI is complex involving signaling through multiple cascades, including lipid peroxidation. Oxidized free fatty acids, a prominent product of lipid peroxidation, are potent cellular mediators involved in induction and resolution of inflammation and modulation of vasomotor tone. While previous studies have assessed lipid peroxidation after TBI, to our knowledge no studies have used a systematic approach to quantify the global oxidative changes in free fatty acids. In this study, we identified and quantified 244 free fatty acid oxidation products using a newly developed global liquid chromatography tandem-mass spectrometry (LC-MS/MS) method. This methodology was used to follow the time course of these lipid species in the contusional cortex of our pediatric rat model of TBI. We show that oxidation peaked at 1h after controlled cortical impact and was progressively attenuated at 4 and 24h time points. While enzymatic and non-enzymatic pathways were activated at 1h post-TBI, enzymatic lipid peroxidation was the predominant mechanism with 15-lipoxygenase (LOX) contributing to the majority of total oxidized fatty acid content. Pro-inflammatory lipid mediators were significantly increased at 1 and 4h after TBI with return to basal levels by 24h. Anti-inflammatory lipid mediators remained significantly increased across all three time points, indicating an elevated and sustained anti-inflammatory response following TBI.
Collapse
Affiliation(s)
- Tamil S Anthonymuthu
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15261, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, United States; Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA 15219, United States
| | - Elizabeth M Kenny
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15261, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, United States; Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA 15219, United States
| | - Andrew A Amoscato
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15219, United States
| | - Jesse Lewis
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15261, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, United States; Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA 15219, United States
| | - Patrick M Kochanek
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15261, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, United States; Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA 15224, United States
| | - Valerian E Kagan
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA 15219, United States; Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15219, United States
| | - Hülya Bayır
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15261, United States; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, United States; Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA 15219, United States; Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15219, United States; Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, PA 15224, United States.
| |
Collapse
|
137
|
Urban A, Golgher L, Brunner C, Gdalyahu A, Har-Gil H, Kain D, Montaldo G, Sironi L, Blinder P. Understanding the neurovascular unit at multiple scales: Advantages and limitations of multi-photon and functional ultrasound imaging. Adv Drug Deliv Rev 2017; 119:73-100. [PMID: 28778714 DOI: 10.1016/j.addr.2017.07.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 07/17/2017] [Accepted: 07/22/2017] [Indexed: 02/07/2023]
Abstract
Developing efficient brain imaging technologies by combining a high spatiotemporal resolution and a large penetration depth is a key step for better understanding the neurovascular interface that emerges as a main pathway to neurodegeneration in many pathologies such as dementia. This review focuses on the advances in two complementary techniques: multi-photon laser scanning microscopy (MPLSM) and functional ultrasound imaging (fUSi). MPLSM has become the gold standard for in vivo imaging of cellular dynamics and morphology, together with cerebral blood flow. fUSi is an innovative imaging modality based on Doppler ultrasound, capable of recording vascular brain activity over large scales (i.e., tens of cubic millimeters) at unprecedented spatial and temporal resolution for such volumes (up to 10μm pixel size at 10kHz). By merging these two technologies, researchers may have access to a more detailed view of the various processes taking place at the neurovascular interface. MPLSM and fUSi are also good candidates for addressing the major challenge of real-time delivery, monitoring, and in vivo evaluation of drugs in neuronal tissue.
Collapse
Affiliation(s)
- Alan Urban
- Neuroelectronics Research Flanders, Leuven, Belgium; VIB, Leuven, Belgium and/or IMEC, Leuven, Belgium; Department of Neurosciences, KU Leuven, Leuven, Belgium; Neurobiology Dept., Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Lior Golgher
- Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| | - Clément Brunner
- Neuroelectronics Research Flanders, Leuven, Belgium; VIB, Leuven, Belgium and/or IMEC, Leuven, Belgium
| | - Amos Gdalyahu
- Neurobiology Dept., Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Hagai Har-Gil
- Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| | - David Kain
- Neurobiology Dept., Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Gabriel Montaldo
- Neuroelectronics Research Flanders, Leuven, Belgium; VIB, Leuven, Belgium and/or IMEC, Leuven, Belgium
| | - Laura Sironi
- Physics Dept., Universita degli Studi di Milano Bicocca, Italy
| | - Pablo Blinder
- Neurobiology Dept., Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel; Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
138
|
Mulherkar S, Firozi K, Huang W, Uddin MD, Grill RJ, Costa-Mattioli M, Robertson C, Tolias KF. RhoA-ROCK Inhibition Reverses Synaptic Remodeling and Motor and Cognitive Deficits Caused by Traumatic Brain Injury. Sci Rep 2017; 7:10689. [PMID: 28878396 PMCID: PMC5587534 DOI: 10.1038/s41598-017-11113-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 08/18/2017] [Indexed: 01/07/2023] Open
Abstract
Traumatic brain injury (TBI) causes extensive neural damage, often resulting in long-term cognitive impairments. Unfortunately, effective treatments for TBI remain elusive. The RhoA-ROCK signaling pathway is a potential therapeutic target since it is activated by TBI and can promote the retraction of dendritic spines/synapses, which are critical for information processing and memory storage. To test this hypothesis, RhoA-ROCK signaling was blocked by RhoA deletion from postnatal neurons or treatment with the ROCK inhibitor fasudil. We found that TBI impairs both motor and cognitive performance and inhibiting RhoA-ROCK signaling alleviates these deficits. Moreover, RhoA-ROCK inhibition prevents TBI-induced spine remodeling and mature spine loss. These data argue that TBI elicits pathological spine remodeling that contributes to behavioral deficits by altering synaptic connections, and RhoA-ROCK inhibition enhances functional recovery by blocking this detrimental effect. As fasudil has been safely used in humans, our results suggest that it could be repurposed to treat TBI.
Collapse
Affiliation(s)
- Shalaka Mulherkar
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Karen Firozi
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Wei Huang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA.,The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, 733N. Broadway, Baltimore, MD, 21205, USA
| | | | - Raymond J Grill
- Department of Integrative Biology and Pharmacology, University of Texas Medical School at Houston, Houston, TX, 77030, USA.,Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Mauro Costa-Mattioli
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA.,Memory and Brain Research Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Claudia Robertson
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kimberley F Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA. .,Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
139
|
Tiran E, Ferrier J, Deffieux T, Gennisson JL, Pezet S, Lenkei Z, Tanter M. Transcranial Functional Ultrasound Imaging in Freely Moving Awake Mice and Anesthetized Young Rats without Contrast Agent. ULTRASOUND IN MEDICINE & BIOLOGY 2017; 43:1679-1689. [PMID: 28476311 PMCID: PMC5754333 DOI: 10.1016/j.ultrasmedbio.2017.03.011] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/07/2017] [Accepted: 03/20/2017] [Indexed: 05/14/2023]
Abstract
Functional ultrasound (fUS) imaging by ultrasensitive Doppler detection of blood volume was previously reported to measure adult rat brain activation and functional connectivity with unmatched spatiotemporal sampling (100 μm, 1 ms), but skull-induced attenuation of ultrasonic waves imposed skull surgery or contrast agent use. Also, fUS feasibility remains to be validated in mice, a major pre-clinical model organism. In the study described here, we performed full-depth ultrasensitive Doppler imaging and 3-D Doppler tomography of the entire mouse brain under anesthesia, non-invasively through the intact skull and skin, without contrast agents. Similar results were obtained in anesthetized young rats up to postnatal day 35, thus enabling longitudinal studies on postnatal brain development. Using a newly developed ultralight ultrasonic probe and an optimized ultrasonic sequence, we also performed minimally invasive full-transcranial fUS imaging of brain vasculature and whisker stimulation-induced barrel cortex activation in awake and freely moving mice, validating transcranial fUS for brain imaging, without anesthesia-induced bias, for behavioral studies.
Collapse
Affiliation(s)
- Elodie Tiran
- INSERM U979, Paris, France; Institut Langevin, CNRS UMR 7587, Paris, France; ESPCI Paris, Paris, France; PSL Research University, Paris, France
| | - Jérémy Ferrier
- ESPCI Paris, Paris, France; PSL Research University, Paris, France; Brain Plasticity Unit, CNRS UMR 8249, Paris, France
| | - Thomas Deffieux
- INSERM U979, Paris, France; Institut Langevin, CNRS UMR 7587, Paris, France; ESPCI Paris, Paris, France; PSL Research University, Paris, France
| | - Jean-Luc Gennisson
- INSERM U979, Paris, France; Institut Langevin, CNRS UMR 7587, Paris, France; ESPCI Paris, Paris, France; PSL Research University, Paris, France
| | - Sophie Pezet
- ESPCI Paris, Paris, France; PSL Research University, Paris, France; Brain Plasticity Unit, CNRS UMR 8249, Paris, France
| | - Zsolt Lenkei
- ESPCI Paris, Paris, France; PSL Research University, Paris, France; Brain Plasticity Unit, CNRS UMR 8249, Paris, France
| | - Mickaël Tanter
- INSERM U979, Paris, France; Institut Langevin, CNRS UMR 7587, Paris, France; ESPCI Paris, Paris, France; PSL Research University, Paris, France.
| |
Collapse
|
140
|
Validation of Acoustic Wave Induced Traumatic Brain Injury in Rats. Brain Sci 2017; 7:brainsci7060059. [PMID: 28574429 PMCID: PMC5483632 DOI: 10.3390/brainsci7060059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 05/17/2017] [Accepted: 05/25/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND This study looked to validate the acoustic wave technology of the Storz-D-Actor that inflicted a consistent closed-head, traumatic brain injury (TBI) in rats. We studied a range of single pulse pressures administered to the rats and observed the resulting decline in motor skills and memory. Histology was observed to measure and confirm the injury insult. METHODS Four different acoustic wave pressures were studied using a single pulse: 0, 3.4, 4.2 and 5.0 bar (n = 10 rats per treatment group). The pulse was administered to the left frontal cortex. Rotarod tests were used to monitor the rats' motor skills while the water maze test was used to monitor memory deficits. The rats were then sacrificed ten days post-treatment for histological analysis of TBI infarct size. RESULTS The behavioral tests showed that acoustic wave technology administered an effective insult causing significant decreases in motor abilities and memory. Histology showed dose-dependent damage to the cortex infarct areas only. CONCLUSIONS This study illustrates that the Storz D-Actor effectively induces a repeatable TBI infarct, avoiding the invasive procedure of a craniotomy often used in TBI research.
Collapse
|
141
|
Mountney A, Boutté AM, Cartagena CM, Flerlage WF, Johnson WD, Rho C, Lu XC, Yarnell A, Marcsisin S, Sousa J, Vuong C, Zottig V, Leung LY, Deng-Bryant Y, Gilsdorf J, Tortella FC, Shear DA. Functional and Molecular Correlates after Single and Repeated Rat Closed-Head Concussion: Indices of Vulnerability after Brain Injury. J Neurotrauma 2017; 34:2768-2789. [PMID: 28326890 DOI: 10.1089/neu.2016.4679] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Closed-head concussive injury is one of the most common causes of traumatic brain injury (TBI). Isolated concussions frequently produce acute neurological impairments, and individuals typically recover spontaneously within a short time frame. In contrast, brain injuries resulting from multiple concussions can result in cumulative damage and elevated risk of developing chronic brain pathologies. Increased attention has focused on identification of diagnostic markers that can prognostically serve as indices of brain health after injury, revealing the temporal profile of vulnerability to a second insult. Such markers may demarcate adequate recovery periods before concussed patients can return to required activities. We developed a noninvasive closed-head impact model that captures the hallmark symptoms of concussion in the absence of gross tissue damage. Animals were subjected to single or repeated concussive impact and examined using a battery of neurological, vestibular, sensorimotor, and molecular metrics. A single concussion induced transient, but marked, acute neurological impairment, gait alterations, neuronal death, and increased glial fibrillary acidic protein (GFAP) expression in brain tissue. As expected, repeated concussions exacerbated sensorimotor dysfunction, prolonged gait abnormalities, induced neuroinflammation, and upregulated GFAP and tau. These animals also exhibited chronic functional neurological impairments with sustained astrogliosis and white matter thinning. Acute changes in molecular signatures correlated with behavioral impairments, whereas increased times to regaining consciousness and balance impairments were associated with higher GFAP and neuroinflammation. Overall, behavioral consequences of either single or repeated concussive impact injuries appeared to resolve more quickly than the underlying molecular, metabolic, and neuropathological abnormalities. This observation, which is supported by similar studies in other mTBI models, underscores the critical need to develop more objective prognostic measures for guiding return-to-play decisions.
Collapse
Affiliation(s)
- Andrea Mountney
- 1 Brain Trauma Neuroprotection and Neurorestoration Branch, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - Angela M Boutté
- 1 Brain Trauma Neuroprotection and Neurorestoration Branch, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - Casandra M Cartagena
- 1 Brain Trauma Neuroprotection and Neurorestoration Branch, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - William F Flerlage
- 1 Brain Trauma Neuroprotection and Neurorestoration Branch, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - Wyane D Johnson
- 1 Brain Trauma Neuroprotection and Neurorestoration Branch, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - Chanyang Rho
- 1 Brain Trauma Neuroprotection and Neurorestoration Branch, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - Xi-Chu Lu
- 1 Brain Trauma Neuroprotection and Neurorestoration Branch, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - Angela Yarnell
- 2 Behavioral Biology Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - Sean Marcsisin
- 3 Division of Experimental Therapeutics, Military Malaria Research, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - Jason Sousa
- 3 Division of Experimental Therapeutics, Military Malaria Research, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - Chau Vuong
- 3 Division of Experimental Therapeutics, Military Malaria Research, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - Victor Zottig
- 3 Division of Experimental Therapeutics, Military Malaria Research, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - Lai-Yee Leung
- 1 Brain Trauma Neuroprotection and Neurorestoration Branch, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - Ying Deng-Bryant
- 1 Brain Trauma Neuroprotection and Neurorestoration Branch, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - Janice Gilsdorf
- 1 Brain Trauma Neuroprotection and Neurorestoration Branch, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - Frank C Tortella
- 1 Brain Trauma Neuroprotection and Neurorestoration Branch, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - Deborah A Shear
- 1 Brain Trauma Neuroprotection and Neurorestoration Branch, Walter Reed Army Institute of Research , Silver Spring, Maryland
| |
Collapse
|
142
|
Brabazon F, Wilson CM, Shukla DK, Mathur S, Jaiswal S, Bermudez S, Byrnes KR, Selwyn R. [18F]FDG-PET Combined with MRI Elucidates the Pathophysiology of Traumatic Brain Injury in Rats. J Neurotrauma 2017; 34:1074-1085. [DOI: 10.1089/neu.2016.4540] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Fiona Brabazon
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Colin M. Wilson
- Department of Radiology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Dinesh K Shukla
- Department of Radiology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Sanjeev Mathur
- Department of Radiology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Shalini Jaiswal
- Department of Radiology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Sara Bermudez
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Kimberly R. Byrnes
- Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Reed Selwyn
- Department of Radiology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Department of Radiology, University of New Mexico, Albuquerque, New Mexico
| |
Collapse
|
143
|
Klee R, Brandt C, Töllner K, Löscher W. Various modifications of the intrahippocampal kainate model of mesial temporal lobe epilepsy in rats fail to resolve the marked rat-to-mouse differences in type and frequency of spontaneous seizures in this model. Epilepsy Behav 2017; 68:129-140. [PMID: 28167446 DOI: 10.1016/j.yebeh.2016.11.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/18/2016] [Accepted: 11/21/2016] [Indexed: 11/19/2022]
Abstract
Temporal lobe epilepsy (TLE) is the most common type of acquired epilepsy in adults. TLE can develop after diverse brain insults, including traumatic brain injury, infections, stroke, or prolonged status epilepticus (SE). Post-SE rodent models of TLE are widely used to understand mechanisms of epileptogenesis and develop treatments for epilepsy prevention. In this respect, the intrahippocampal kainate model of TLE in mice is of interest, because highly frequent spontaneous electrographic seizures develop in the kainate focus, allowing evaluation of both anti-seizure and anti-epileptogenic effects of novel drugs with only short EEG recording periods, which is not possible in any other model of TLE, including the intrahippocampal kainate model in rats. In the present study, we investigated whether the marked mouse-to-rat difference in occurrence and frequency of spontaneous seizures is due to a species difference or to technical variables, such as anesthesia during kainate injection, kainate dose, or location of kainate injection and EEG electrode in the hippocampus. When, as in the mouse model, anesthesia was used during kainate injection, only few rats developed epilepsy, although severity or duration of SE was not affected by isoflurane. In contrast, most rats developed epilepsy when kainate was injected without anesthesia. However, frequent electrographic seizures as observed in mice did not occur in rats, irrespective of location of kainate injection (CA1, CA3) or EEG recording electrode (CA1, CA3, dentate gyrus) or dose of kainate injected. These data indicate marked phenotypic differences between mice and rats in this model. Further studies should explore the mechanisms underlying this species difference.
Collapse
Affiliation(s)
- Rebecca Klee
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany, and Center for Systems Neuroscience, Hannover, Germany
| | - Claudia Brandt
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany, and Center for Systems Neuroscience, Hannover, Germany
| | - Kathrin Töllner
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany, and Center for Systems Neuroscience, Hannover, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany, and Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
144
|
Kononenko O, Galatenko V, Andersson M, Bazov I, Watanabe H, Zhou XW, Iatsyshyna A, Mityakina I, Yakovleva T, Sarkisyan D, Ponomarev I, Krishtal O, Marklund N, Tonevitsky A, Adkins DL, Bakalkin G. Intra- and interregional coregulation of opioid genes: broken symmetry in spinal circuits. FASEB J 2017; 31:1953-1963. [PMID: 28122917 DOI: 10.1096/fj.201601039r] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 01/09/2017] [Indexed: 12/31/2022]
Abstract
Regulation of the formation and rewiring of neural circuits by neuropeptides may require coordinated production of these signaling molecules and their receptors that may be established at the transcriptional level. Here, we address this hypothesis by comparing absolute expression levels of opioid peptides with their receptors, the largest neuropeptide family, and by characterizing coexpression (transcriptionally coordinated) patterns of these genes. We demonstrated that expression patterns of opioid genes highly correlate within and across functionally and anatomically different areas. Opioid peptide genes, compared with their receptor genes, are transcribed at much greater absolute levels, which suggests formation of a neuropeptide cloud that covers the receptor-expressed circuits. Surprisingly, we found that both expression levels and the proportion of opioid receptors are strongly lateralized in the spinal cord, interregional coexpression patterns are side specific, and intraregional coexpression profiles are affected differently by left- and right-side unilateral body injury. We propose that opioid genes are regulated as interconnected components of the same molecular system distributed between distinct anatomic regions. The striking feature of this system is its asymmetric coexpression patterns, which suggest side-specific regulation of selective neural circuits by opioid neurohormones.-Kononenko, O., Galatenko, V., Andersson, M., Bazov, I., Watanabe, H., Zhou, X. W., Iatsyshyna, A., Mityakina, I., Yakovleva, T., Sarkisyan, D., Ponomarev, I., Krishtal, O., Marklund, N., Tonevitsky, A., Adkins, D. L., Bakalkin, G. Intra- and interregional coregulation of opioid genes: broken symmetry in spinal circuits.
Collapse
Affiliation(s)
- Olga Kononenko
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.,Key State Laboratory, Bogomoletz Institute of Physiology, Kiev, Ukraine
| | | | - Malin Andersson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Igor Bazov
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden;
| | - Hiroyuki Watanabe
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Xing Wu Zhou
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Anna Iatsyshyna
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.,Department of Human Genetics, Institute of Molecular Biology and Genetics, Kiev, Ukraine
| | | | - Tatiana Yakovleva
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Daniil Sarkisyan
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Igor Ponomarev
- Waggoner Center for Alcohol and Addiction Research and The College of Pharmacy, The University of Texas, Austin, Texas, USA
| | - Oleg Krishtal
- Key State Laboratory, Bogomoletz Institute of Physiology, Kiev, Ukraine
| | - Niklas Marklund
- Department of Neuroscience, Section of Neurosurgery, Uppsala University Hospital, Uppsala, Sweden
| | | | - DeAnna L Adkins
- Department of Neuroscience, College of Medicine, and.,Department of Health Sciences and Research, College of Health Professions, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Georgy Bakalkin
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
145
|
Brunner C, Isabel C, Martin A, Dussaux C, Savoye A, Emmrich J, Montaldo G, Mas JL, Baron JC, Urban A. Mapping the dynamics of brain perfusion using functional ultrasound in a rat model of transient middle cerebral artery occlusion. J Cereb Blood Flow Metab 2017; 37:263-276. [PMID: 26721392 PMCID: PMC5363744 DOI: 10.1177/0271678x15622466] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 11/17/2015] [Accepted: 11/18/2015] [Indexed: 01/07/2023]
Abstract
Following middle cerebral artery occlusion, tissue outcome ranges from normal to infarcted depending on depth and duration of hypoperfusion as well as occurrence and efficiency of reperfusion. However, the precise time course of these changes in relation to tissue and behavioral outcome remains unsettled. To address these issues, a three-dimensional wide field-of-view and real-time quantitative functional imaging technique able to map perfusion in the rodent brain would be desirable. Here, we applied functional ultrasound imaging, a novel approach to map relative cerebral blood volume without contrast agent, in a rat model of brief proximal transient middle cerebral artery occlusion to assess perfusion in penetrating arterioles and venules acutely and over six days thanks to a thinned-skull preparation. Functional ultrasound imaging efficiently mapped the acute changes in relative cerebral blood volume during occlusion and following reperfusion with high spatial resolution (100 µm), notably documenting marked focal decreases during occlusion, and was able to chart the fine dynamics of tissue reperfusion (rate: one frame/5 s) in the individual rat. No behavioral and only mild post-mortem immunofluorescence changes were observed. Our study suggests functional ultrasound is a particularly well-adapted imaging technique to study cerebral perfusion in acute experimental stroke longitudinally from the hyper-acute up to the chronic stage in the same subject.
Collapse
Affiliation(s)
- Clément Brunner
- Stroke Research Group, Centre de Psychiatrie et Neuroscience, INSERM U894, Hôpital Sainte-Anne, Paris, France.,SANOFI Research and Development, Lead Generation to Candidate Realization, Chilly-Mazarin, France
| | - Clothilde Isabel
- Stroke Research Group, Centre de Psychiatrie et Neuroscience, INSERM U894, Hôpital Sainte-Anne, Paris, France
| | - Abraham Martin
- Molecular Imaging Unit, CIC biomaGUNE, San Sebastián, Spain
| | - Clara Dussaux
- Stroke Research Group, Centre de Psychiatrie et Neuroscience, INSERM U894, Hôpital Sainte-Anne, Paris, France
| | - Anne Savoye
- Stroke Research Group, Centre de Psychiatrie et Neuroscience, INSERM U894, Hôpital Sainte-Anne, Paris, France
| | | | - Gabriel Montaldo
- Stroke Research Group, Centre de Psychiatrie et Neuroscience, INSERM U894, Hôpital Sainte-Anne, Paris, France
| | - Jean-Louis Mas
- Stroke Research Group, Centre de Psychiatrie et Neuroscience, INSERM U894, Hôpital Sainte-Anne, Paris, France
| | - Jean-Claude Baron
- Stroke Research Group, Centre de Psychiatrie et Neuroscience, INSERM U894, Hôpital Sainte-Anne, Paris, France
| | - Alan Urban
- Stroke Research Group, Centre de Psychiatrie et Neuroscience, INSERM U894, Hôpital Sainte-Anne, Paris, France
| |
Collapse
|
146
|
Mondello S, Shear DA, Bramlett HM, Dixon CE, Schmid KE, Dietrich WD, Wang KKW, Hayes RL, Glushakova O, Catania M, Richieri SP, Povlishock JT, Tortella FC, Kochanek PM. Insight into Pre-Clinical Models of Traumatic Brain Injury Using Circulating Brain Damage Biomarkers: Operation Brain Trauma Therapy. J Neurotrauma 2016; 33:595-605. [PMID: 26671651 DOI: 10.1089/neu.2015.4132] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Operation Brain Trauma Therapy (OBTT) is a multicenter pre-clinical drug screening consortium testing promising therapies for traumatic brain injury (TBI) in three well-established models of TBI in rats--namely, parasagittal fluid percussion injury (FPI), controlled cortical impact (CCI), and penetrating ballistic-like brain injury (PBBI). This article presents unique characterization of these models using histological and behavioral outcomes and novel candidate biomarkers from the first three treatment trials of OBTT. Adult rats underwent CCI, FPI, or PBBI and were treated with vehicle (VEH). Shams underwent all manipulations except trauma. The glial marker glial fibrillary acidic protein (GFAP) and the neuronal marker ubiquitin C-terminal hydrolase (UCH-L1) were measured by enzyme-linked immunosorbent assay in blood at 4 and 24 h, and their delta 24-4 h was calculated for each marker. Comparing sham groups across experiments, no differences were found in the same model. Similarly, comparing TBI + VEH groups across experiments, no differences were found in the same model. GFAP was acutely increased in injured rats in each model, with significant differences in levels and temporal patterns mirrored by significant differences in delta 24-4 h GFAP levels and neuropathological and behavioral outcomes. Circulating GFAP levels at 4 and 24 h were powerful predictors of 21 day contusion volume and tissue loss. UCH-L1 showed similar tendencies, albeit with less robust differences between sham and injury groups. Significant differences were also found comparing shams across the models. Our findings (1) demonstrate that TBI models display specific biomarker profiles, functional deficits, and pathological consequence; (2) support the concept that there are different cellular, molecular, and pathophysiological responses to TBI in each model; and (3) advance our understanding of TBI, providing opportunities for a successful translation and holding promise for theranostic applications. Based on our findings, additional studies in pre-clinical models should pursue assessment of GFAP as a surrogate histological and/or theranostic end-point.
Collapse
Affiliation(s)
- Stefania Mondello
- 1 Department of Neurosciences, University of Messina , Messina, Italy
| | - Deborah A Shear
- 2 Brain Trauma Neuroprotection/Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - Helen M Bramlett
- 3 Department of Neurological Surgery, The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami , Miami, Florida.,4 Bruce W. Carter Department of Veterans Affairs Medical Center , Miami, Florida
| | - C Edward Dixon
- 5 Department of Neurological Surgery, Brain Trauma Research Center, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Kara E Schmid
- 2 Brain Trauma Neuroprotection/Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - W Dalton Dietrich
- 3 Department of Neurological Surgery, The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami , Miami, Florida
| | - Kevin K W Wang
- 6 Center of Neuroproteomics and Biomarkers Research, Department of Psychiatry and Neuroscience, University of Florida , Gainesville, Florida
| | - Ronald L Hayes
- 7 Center for Innovative Research, Center for Neuroproteomics and Biomarkers Research , Banyan Biomarkers, Inc., Alachua, Florida
| | | | | | | | - John T Povlishock
- 9 Department of Anatomy and Neurobiology, Virginia Commonwealth University , Richmond, Virginia
| | - Frank C Tortella
- 2 Brain Trauma Neuroprotection/Neurorestoration, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research , Silver Spring, Maryland
| | - Patrick M Kochanek
- 10 Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| |
Collapse
|
147
|
Haar CV, Lam FC, Adams WA, Riparip LK, Kaur S, Muthukrishna M, Rosi S, Winstanley CA. Frontal Traumatic Brain Injury in Rats Causes Long-Lasting Impairments in Impulse Control That Are Differentially Sensitive to Pharmacotherapeutics and Associated with Chronic Neuroinflammation. ACS Chem Neurosci 2016; 7:1531-1542. [PMID: 27525447 PMCID: PMC9487719 DOI: 10.1021/acschemneuro.6b00166] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Traumatic brain injury (TBI) affects millions yearly, and is increasingly associated with chronic neuropsychiatric symptoms. We assessed the long-term effects of different bilateral frontal controlled cortical impact injury severities (mild, moderate, and severe) on the five-choice serial reaction time task, a paradigm with relatively independent measurements of attention, motor impulsivity, and motivation. Moderately- and severely injured animals exhibited impairments across all cognitive domains that were still evident 14 weeks postinjury, while mild-injured animals only demonstrated persistent deficits in impulse control. However, recovery of function varied considerably between subjects such that some showed no impairment ("TBI-resilient"), some demonstrated initial deficits that recovered ("TBI-vulnerable"), and some never recovered ("chronically-impaired"). Three clinically relevant treatments for impulse-control or TBI, amphetamine, atomoxetine, and amantadine, were assessed for efficacy in treating injury-induced deficits. Susceptibility to TBI affected the response to pharmacological challenge with amphetamine. Whereas sham and TBI-resilient animals showed characteristic impairments in impulse control at higher doses, amphetamine had the opposite effect in chronically impaired rats, improving task performance. In contrast, atomoxetine and amantadine reduced premature responding but increased omissions, suggesting psychomotor slowing. Analysis of brain tissue revealed that generalized neuroinflammation was associated with impulsivity even when accounting for the degree of brain damage. This is one of the first studies to characterize psychiatric-like symptoms in experimental TBI. Our data highlight the importance of testing pharmacotherapies in TBI models in order to predict efficacy, and suggest that neuroinflammation may represent a treatment target for impulse control problems following injury.
Collapse
Affiliation(s)
- Cole Vonder Haar
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada,Department of Psychology, University of British Columbia, Vancouver, BC, Canada,Corresponding author: Cole Vonder Haar or Catharine A. Winstanley, CV: Department of Psychology, 53 Campus Dr, Morgantown, WV, 26506, Tel: 1-304-293-1787, , CAW: Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada, Tel: 1-604-822-2024,
| | - Frederick C.W. Lam
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada,Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Wendy A. Adams
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada,Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Lara-Kirstie Riparip
- Brain and Spinal Injury Center, Departments of Physical Therapy Rehabilitation Science and Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Sukhbir Kaur
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | | | - Susanna Rosi
- Brain and Spinal Injury Center, Departments of Physical Therapy Rehabilitation Science and Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Catharine A. Winstanley
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada,Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
148
|
GSNO promotes functional recovery in experimental TBI by stabilizing HIF-1α. Behav Brain Res 2016; 340:63-70. [PMID: 27780722 DOI: 10.1016/j.bbr.2016.10.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 10/01/2016] [Accepted: 10/21/2016] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) causes sustained disability due to compromised neurorepair mechanisms. Crucial to neurorepair and functional recovery following both TBI and stroke is hypoxia-inducible factor-1 alpha (HIF-1α). Based on reports that HIF-1α could be stabilized via S-nitrosylation, we tested the hypothesis that the S-nitrosylating agent S-nitrosoglutathione (GSNO) would stabilize HIF-1α, thereby stimulating neurorepair mechanisms and aiding in functional recovery. TBI was induced by controlled cortical impact (CCI) in adult rats. GSNO (0.05mg/kg) was administered at two hours after CCI. The treatment was repeated daily until the 14th day after CCI. Functional recovery was assessed by motor and cognitive functions, and the recovery was compared with the expression of HIF-1α. The mechanisms of GSNO-mediated S-nitrosylation of HIF-1α were determined using brain endothelial cells. While non-treated TBI animals showed sustained neurobehavioral deficits, GSNO treatment of TBI improved neurobehavioral functions. GSNO also increased the expression of HIF-1α and VEGF. The beneficial effects of GSNO on neurobehavioral functions in TBI animals were blocked by treatment with the HIF-1α inhibitor 2-methoxyestradiol (2-ME). The stimulatory effect of GSNO on VEGF was reversed not only by 2-ME but also by the denitrosylating agent dithiothreitol, confirming our hypothesis that GSNO's benefits are mediated by the stabilization of HIF-1α via S-nitrosylation. GSNO's S-nitrosylation of HIF-1α was further confirmed using a biotin switch assay in endothelial cells. The data provide evidence that GSNO treatment of TBI aids functional recovery through stabilizing HIF-1α via S-nitrosylation. GSNO is a natural component of the human brain/body, and its exogenous administration has not shown adverse effects in humans. Therefore, the translational potential of GSNO therapy in TBI is high.
Collapse
|
149
|
Grin’kina NM, Li Y, Haber M, Sangobowale M, Nikulina E, Le’Pre C, El Sehamy AM, Dugue R, Ho JS, Bergold PJ. Righting Reflex Predicts Long-Term Histological and Behavioral Outcomes in a Closed Head Model of Traumatic Brain Injury. PLoS One 2016; 11:e0161053. [PMID: 27657499 PMCID: PMC5033343 DOI: 10.1371/journal.pone.0161053] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 07/30/2016] [Indexed: 12/22/2022] Open
Abstract
Blunt impact produces a heterogeneous brain injury in people and in animal models of traumatic brain injury. We report that a single closed head impact to adult C57/BL6 mice produced two injury syndromes (CHI-1 and CHI-2). CHI-1 mice spontaneously reinitiated breathing after injury while CHI-2 mice had prolonged apnea and regained breathing only after cardiopulmonary resuscitation and supplementation of 100% O2. The CHI-1 group significantly regained righting reflex more rapidly than the CHI-2 group. At 7 days post-injury, CHI-1, but not CHI-2 mice, acquired but had no long-term retention of an active place avoidance task. The behavioral deficits of CHI-1 and CHI-2 mice were retained one-month after the injury. CHI-1 mice had loss of hippocampal neurons and localized white matter injury at one month after injury. CHI-2 had a larger loss of hippocampal neurons and more widespread loss of myelin and axons. High-speed videos made during the injury were followed by assessment of breathing and righting reflex. These videos show that CHI-2 mice experienced a larger vertical g-force than CHI-1 mice. Time to regain righting reflex in CHI-2 mice significantly correlated with vertical g-force. Thus, physiological responses occurring immediately after injury can be valuable surrogate markers of subsequent behavioral and histological deficits.
Collapse
Affiliation(s)
- Natalia M. Grin’kina
- Robert F. Furchgott Center for Neural Science, SUNY-Downstate Medical Center, Brooklyn, NY, United States of America
- Department of Physiology and Pharmacology SUNY-Downstate Medical Center, 450 Clarkson Avenue, Brooklyn, NY, 11203, United States of America
| | - Yang Li
- Robert F. Furchgott Center for Neural Science, SUNY-Downstate Medical Center, Brooklyn, NY, United States of America
| | - Margalit Haber
- Program in Neural and Behavioral Science, SUNY-Downstate Medical Center, Brooklyn, NY, United States of America
- Robert F. Furchgott Center for Neural Science, SUNY-Downstate Medical Center, Brooklyn, NY, United States of America
| | - Michael Sangobowale
- Program in Neural and Behavioral Science, SUNY-Downstate Medical Center, Brooklyn, NY, United States of America
| | - Elena Nikulina
- Robert F. Furchgott Center for Neural Science, SUNY-Downstate Medical Center, Brooklyn, NY, United States of America
- Department of Physiology and Pharmacology SUNY-Downstate Medical Center, 450 Clarkson Avenue, Brooklyn, NY, 11203, United States of America
| | - Charm Le’Pre
- Program in Neural and Behavioral Science, SUNY-Downstate Medical Center, Brooklyn, NY, United States of America
| | - Alexander M. El Sehamy
- Robert F. Furchgott Center for Neural Science, SUNY-Downstate Medical Center, Brooklyn, NY, United States of America
| | - Rachelle Dugue
- Program in Neural and Behavioral Science, SUNY-Downstate Medical Center, Brooklyn, NY, United States of America
| | - Johnson S. Ho
- Robert F. Furchgott Center for Neural Science, SUNY-Downstate Medical Center, Brooklyn, NY, United States of America
| | - Peter J. Bergold
- Program in Neural and Behavioral Science, SUNY-Downstate Medical Center, Brooklyn, NY, United States of America
- Robert F. Furchgott Center for Neural Science, SUNY-Downstate Medical Center, Brooklyn, NY, United States of America
- Department of Physiology and Pharmacology SUNY-Downstate Medical Center, 450 Clarkson Avenue, Brooklyn, NY, 11203, United States of America
- * E-mail:
| |
Collapse
|
150
|
Shultz SR, McDonald SJ, Vonder Haar C, Meconi A, Vink R, van Donkelaar P, Taneja C, Iverson GL, Christie BR. The potential for animal models to provide insight into mild traumatic brain injury: Translational challenges and strategies. Neurosci Biobehav Rev 2016; 76:396-414. [PMID: 27659125 DOI: 10.1016/j.neubiorev.2016.09.014] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 09/07/2016] [Accepted: 09/16/2016] [Indexed: 12/14/2022]
Abstract
Mild traumatic brain injury (mTBI) is a common health problem. There is tremendous variability and heterogeneity in human mTBI, including mechanisms of injury, biomechanical forces, injury severity, spatial and temporal pathophysiology, genetic factors, pre-injury vulnerability and resilience factors, and clinical outcomes. Animal models greatly reduce this variability and heterogeneity, and provide a means to study mTBI in a rigorous, controlled, and efficient manner. Rodent models, in particular, are time- and cost-efficient, and they allow researchers to measure morphological, cellular, molecular, and behavioral variables in a single study. However, inter-species differences in anatomy, morphology, metabolism, neurobiology, and lifespan create translational challenges. Although the term "mild" TBI is used often in the pre-clinical literature, clearly defined criteria for mild, moderate, and severe TBI in animal models have not been agreed upon. In this review, we introduce current issues facing the mTBI field, summarize the available research methodologies and previous studies in mTBI animal models, and discuss how a translational research approach may be useful in advancing our understanding and management of mTBI.
Collapse
Affiliation(s)
- Sandy R Shultz
- Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia.
| | - Stuart J McDonald
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Cole Vonder Haar
- Department of Psychology, The University of British Columbia, Vancouver, BC, Canada
| | - Alicia Meconi
- Division of Medical Sciences, The University of Victoria, Victoria, BC, Canada
| | - Robert Vink
- Division of Health Sciences, The University of South Australia, Adelaide, SA, Australia
| | - Paul van Donkelaar
- School of Health and Exercise Sciences, The University of British Columbia Okanagan, Kelowna, BC, Canada
| | - Chand Taneja
- Division of Medical Sciences, The University of Victoria, Victoria, BC, Canada
| | - Grant L Iverson
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Spaulding Rehabilitation Hospital, Home Base, A Red Sox Foundation and Massachusetts General Hospital Program, and MassGeneral Hospital for Children™ Sports Concussion Program, Boston, MA, USA
| | - Brian R Christie
- Division of Medical Sciences, The University of Victoria, Victoria, BC, Canada
| |
Collapse
|