1
|
Treble-Barna A, Petersen BA, Stec Z, Conley YP, Fink EL, Kochanek PM. Brain-Derived Neurotrophic Factor in Pediatric Acquired Brain Injury and Recovery. Biomolecules 2024; 14:191. [PMID: 38397427 PMCID: PMC10886547 DOI: 10.3390/biom14020191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
We review emerging preclinical and clinical evidence regarding brain-derived neurotrophic factor (BDNF) protein, genotype, and DNA methylation (DNAm) as biomarkers of outcomes in three important etiologies of pediatric acquired brain injury (ABI), traumatic brain injury, global cerebral ischemia, and stroke. We also summarize evidence suggesting that BDNF is (1) involved in the biological embedding of the psychosocial environment, (2) responsive to rehabilitative therapies, and (3) potentially modifiable. BDNF's unique potential as a biomarker of neuroplasticity and neural repair that is reflective of and responsive to both pre- and post-injury environmental influences separates it from traditional protein biomarkers of structural brain injury with exciting potential to advance pediatric ABI management by increasing the accuracy of prognostic tools and informing clinical decision making through the monitoring of therapeutic effects.
Collapse
Affiliation(s)
- Amery Treble-Barna
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (B.A.P.); (Z.S.)
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (E.L.F.); (P.M.K.)
| | - Bailey A. Petersen
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (B.A.P.); (Z.S.)
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (E.L.F.); (P.M.K.)
| | - Zachary Stec
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (B.A.P.); (Z.S.)
| | - Yvette P. Conley
- Department of Health Promotion & Development, University of Pittsburgh School of Nursing, Pittsburgh, PA 15213, USA;
| | - Ericka L. Fink
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (E.L.F.); (P.M.K.)
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Patrick M. Kochanek
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (E.L.F.); (P.M.K.)
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
2
|
The Role of BDNF in Experimental and Clinical Traumatic Brain Injury. Int J Mol Sci 2021; 22:ijms22073582. [PMID: 33808272 PMCID: PMC8037220 DOI: 10.3390/ijms22073582] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023] Open
Abstract
Traumatic brain injury is one of the leading causes of mortality and morbidity in the world with no current pharmacological treatment. The role of BDNF in neural repair and regeneration is well established and has also been the focus of TBI research. Here, we review experimental animal models assessing BDNF expression following injury as well as clinical studies in humans including the role of BDNF polymorphism in TBI. There is a large heterogeneity in experimental setups and hence the results with different regional and temporal changes in BDNF expression. Several studies have also assessed different interventions to affect the BDNF expression following injury. Clinical studies highlight the importance of BDNF polymorphism in the outcome and indicate a protective role of BDNF polymorphism following injury. Considering the possibility of affecting the BDNF pathway with available substances, we discuss future studies using transgenic mice as well as iPSC in order to understand the underlying mechanism of BDNF polymorphism in TBI and develop a possible pharmacological treatment.
Collapse
|
3
|
Grovola MR, Paleologos N, Wofford KL, Harris JP, Browne KD, Johnson V, Duda JE, Wolf JA, Cullen DK. Mossy cell hypertrophy and synaptic changes in the hilus following mild diffuse traumatic brain injury in pigs. J Neuroinflammation 2020; 17:44. [PMID: 32005260 PMCID: PMC6993507 DOI: 10.1186/s12974-020-1720-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 01/19/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Each year in the USA, over 2.4 million people experience mild traumatic brain injury (TBI), which can induce long-term neurological deficits. The dentate gyrus of the hippocampus is notably susceptible to damage following TBI, as hilar mossy cell changes in particular may contribute to post-TBI dysfunction. Moreover, microglial activation after TBI may play a role in hippocampal circuit and/or synaptic remodeling; however, the potential effects of chronic microglial changes are currently unknown. The objective of the current study was to assess neuropathological and neuroinflammatory changes in subregions of the dentate gyrus at acute to chronic time points following mild TBI using an established model of closed-head rotational acceleration induced TBI in pigs. METHODS This study utilized archival tissue of pigs which were subjected to sham conditions or rapid head rotation in the coronal plane to generate mild TBI. A quantitative assessment of neuropathological changes in the hippocampus was performed via immunohistochemical labeling of whole coronal tissue sections at 3 days post-injury (DPI), 7 DPI, 30 DPI, and 1 year post-injury (YPI), with a focus on mossy cell atrophy and synaptic reorganization, in context with microglial alterations (e.g., density, proximity to mossy cells) in the dentate gyrus. RESULTS There were no changes in mossy cell density between sham and injured animals, indicating no frank loss of mossy cells at the mild injury level evaluated. However, we found significant mossy cell hypertrophy at 7 DPI and 30 DPI in anterior (> 16% increase in mean cell area at each time; p = < 0.001 each) and 30 DPI in posterior (8.3% increase; p = < 0.0001) hippocampus. We also found dramatic increases in synapsin staining around mossy cells at 7 DPI in both anterior (74.7% increase in synapsin labeling; p = < 0.0001) and posterior (82.7% increase; p = < 0.0001) hippocampus. Interestingly, these morphological and synaptic alterations correlated with a significant change in microglia in proximity to mossy cells at 7 DPI in anterior and at 30 DPI in the posterior hippocampus. For broader context, while we found that there were significant increases in microglia density in the granule cell layer at 30 DPI (anterior and posterior) and 1 YPI (posterior only) and in the molecular layer at 1 YPI (anterior only), we found no significant changes in overall microglial density in the hilus at any of the time points evaluated post-injury. CONCLUSIONS The alterations of mossy cell size and synaptic inputs paired with changes in microglia density around the cells demonstrate the susceptibility of hilar mossy cells after even mild TBI. This subtle hilar mossy cell pathology may play a role in aberrant hippocampal function post-TBI, although additional studies are needed to characterize potential physiological and cognitive alterations.
Collapse
Affiliation(s)
- Michael R Grovola
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, 105E Hayden Hall/3320 Smith Walk, Philadelphia, PA, 19104, USA
| | - Nicholas Paleologos
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, 105E Hayden Hall/3320 Smith Walk, Philadelphia, PA, 19104, USA
| | - Kathryn L Wofford
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, 105E Hayden Hall/3320 Smith Walk, Philadelphia, PA, 19104, USA
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - James P Harris
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, 105E Hayden Hall/3320 Smith Walk, Philadelphia, PA, 19104, USA
| | - Kevin D Browne
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, 105E Hayden Hall/3320 Smith Walk, Philadelphia, PA, 19104, USA
| | - Victoria Johnson
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, 105E Hayden Hall/3320 Smith Walk, Philadelphia, PA, 19104, USA
| | - John E Duda
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Parkinson's Disease Research, Education and Clinical Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John A Wolf
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, 105E Hayden Hall/3320 Smith Walk, Philadelphia, PA, 19104, USA
| | - D Kacy Cullen
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA.
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, 105E Hayden Hall/3320 Smith Walk, Philadelphia, PA, 19104, USA.
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, 105E Hayden Hall/3320 Smith Walk, Philadelphia, PA, 19104, USA.
| |
Collapse
|
4
|
Sudarshan K, Boda AK, Dogra S, Bose I, Yadav PN, Aidhen IS. Discovery of an isocoumarin analogue that modulates neuronal functions via neurotrophin receptor TrkB. Bioorg Med Chem Lett 2019; 29:585-590. [PMID: 30600206 DOI: 10.1016/j.bmcl.2018.12.057] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/22/2018] [Accepted: 12/25/2018] [Indexed: 01/07/2023]
|
5
|
Sullivan KA, Hills AP, Iverson GL. Graded Combined Aerobic Resistance Exercise (CARE) to Prevent or Treat the Persistent Post-concussion Syndrome. Curr Neurol Neurosci Rep 2018; 18:75. [DOI: 10.1007/s11910-018-0884-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
6
|
Chandel S, Gupta SK, Medhi B. Epileptogenesis following experimentally induced traumatic brain injury - a systematic review. Rev Neurosci 2018; 27:329-46. [PMID: 26581067 DOI: 10.1515/revneuro-2015-0050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 10/21/2015] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) is a complex neurotrauma in civilian life and the battlefield with a broad spectrum of symptoms, long-term neuropsychological disability, as well as mortality worldwide. Posttraumatic epilepsy (PTE) is a common outcome of TBI with unknown mechanisms, followed by posttraumatic epileptogenesis. There are numerous rodent models of TBI available with varying pathomechanisms of head injury similar to human TBI, but there is no evidence for an adequate TBI model that can properly mimic all aspects of clinical TBI and the first successive spontaneous focal seizures follow a single episode of neurotrauma with respect to epileptogenesis. This review aims to provide current information regarding the various experimental animal models of TBI relevant to clinical TBI. Mossy fiber sprouting, loss of dentate hilar neurons along with recurrent seizures, and epileptic discharge similar to human PTE have been studied in fluid percussion injury, weight-drop injury, and cortical impact models, but further refinement of animal models and functional test is warranted to better understand the underlying pathophysiology of posttraumatic epileptogenesis. A multifaceted research approach in TBI model may lead to exploration of the potential treatment measures, which are a major challenge to the research community and drug developers. With respect to clinical setting, proper patient data collection, improved clinical trials with advancement in drug delivery strategies, blood-brain barrier permeability, and proper monitoring of level and effects of target drug are also important.
Collapse
|
7
|
Neurotrauma: The Crosstalk between Neurotrophins and Inflammation in the Acutely Injured Brain. Int J Mol Sci 2017; 18:ijms18051082. [PMID: 28524074 PMCID: PMC5454991 DOI: 10.3390/ijms18051082] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 04/25/2017] [Accepted: 05/11/2017] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) is a major cause of morbidity and mortality among young individuals worldwide. Understanding the pathophysiology of neurotrauma is crucial for the development of more effective therapeutic strategies. After the trauma occurs, immediate neurologic damage is produced by the traumatic forces; this primary injury triggers a secondary wave of biochemical cascades together with metabolic and cellular changes, called secondary neural injury. In the scenario of the acutely injured brain, the ongoing secondary injury results in ischemia and edema culminating in an uncontrollable increase in intracranial pressure. These areas of secondary injury progression, or areas of “traumatic penumbra”, represent crucial targets for therapeutic interventions. Neurotrophins are a class of signaling molecules that promote survival and/or maintenance of neurons. They also stimulate axonal growth, synaptic plasticity, and neurotransmitter synthesis and release. Therefore, this review focuses on the role of neurotrophins in the acute post-injury response. Here, we discuss possible endogenous neuroprotective mechanisms of neurotrophins in the prevailing environment surrounding the injured areas, and highlight the crosstalk between neurotrophins and inflammation with focus on neurovascular unit cells, particularly pericytes. The perspective is that neurotrophins may represent promising targets for research on neuroprotective and neurorestorative processes in the short-term following TBI.
Collapse
|
8
|
Corrigan F, Arulsamy A, Teng J, Collins-Praino LE. Pumping the Brakes: Neurotrophic Factors for the Prevention of Cognitive Impairment and Dementia after Traumatic Brain Injury. J Neurotrauma 2016; 34:971-986. [PMID: 27630018 DOI: 10.1089/neu.2016.4589] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is the leading cause of disability and death worldwide, affecting as many as 54,000,000-60,000,000 people annually. TBI is associated with significant impairments in brain function, impacting cognitive, emotional, behavioral, and physical functioning. Although much previous research has focused on the impairment immediately following injury, TBI may have much longer-lasting consequences, including neuropsychiatric disorders and cognitive impairment. TBI, even mild brain injury, has also been recognized as a significant risk factor for the later development of dementia and Alzheimer's disease. Although the link between TBI and dementia is currently unknown, several proposed mechanisms have been put forward, including alterations in glucose metabolism, excitotoxicity, calcium influx, mitochondrial dysfunction, oxidative stress, and neuroinflammation. A treatment for the devastating long-term consequences of TBI is desperately needed. Unfortunately, however, no such treatment is currently available, making this a major area of unmet medical need. Increasing the level of neurotrophic factor expression in key brain areas may be one potential therapeutic strategy. Of the neurotrophic factors, granulocyte-colony stimulating factor (G-CSF) may be particularly effective for preventing the emergence of long-term complications of TBI, including dementia, because of its ability to reduce apoptosis, stimulate neurogenesis, and increase neuroplasticity.
Collapse
Affiliation(s)
- Frances Corrigan
- Translational Neuropathology Lab, Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide , Adelaide, Australia
| | - Alina Arulsamy
- Translational Neuropathology Lab, Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide , Adelaide, Australia
| | - Jason Teng
- Translational Neuropathology Lab, Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide , Adelaide, Australia
| | - Lyndsey E Collins-Praino
- Translational Neuropathology Lab, Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide , Adelaide, Australia
| |
Collapse
|
9
|
Wang TT, Tian C, Sun J, Wang H, Zhang BY, Chen C, Wang J, Xiao K, Chen LN, Lv Y, Gao C, Shi Q, Xin Y, Dong XP. Down-regulation of brain-derived neurotrophic factor and its signaling components in the brain tissues of scrapie experimental animals. Int J Biochem Cell Biol 2016; 79:318-326. [PMID: 27590859 DOI: 10.1016/j.biocel.2016.08.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 08/11/2016] [Accepted: 08/29/2016] [Indexed: 12/30/2022]
Abstract
Prion is a unique nucleic acid-free pathogen that causes human and animal fatal neurodegenerative diseases. Brain-derived neurotrophic factor (BDNF) is a prototypic neurotrophin that helps to support the survival of existing neurons, and encourage the growth and differentiation of new neurons and synapses through axonal and dendritic sprouting. There are two distinct classes of glycosylated receptors, neurotrophin receptor p75 (p75NTR) and tropomyosin-related kinase (Trk), that can bind to BDNF. To obtain insights into the possible alterations of brain BDNF and its signaling pathway in prion disease, the levels of BDNF and several molecules in the BDNF pathway in the brain tissues of scrapie agents 263K-infected hamsters were separately evaluated. Western blots and/or immunohistochemical (IHC) assays revealed that BDNF, TrkB, GRB2 and p75NTR, were significantly downregulated in the brain tissues of scrapie-infected rodents at terminal stage. Double-stained immunofluorescent assay (IFA) demonstrated that BDNF and phospho-TrkB predominately expressed in neurons. Dynamic analyses of the brain samples collected at the different time-points during the incubation period illustrated continuous decreases of BDNF, TrkB, phospho-TrkB, GRB2 and p75NTR, which correlated well with neuron loss. However, these proteins remained almost unchanged in the prion infected cell line SMB-S15 compared with those of its normal cell line SMB-PS. These data suggest that the BDNF signaling pathway is severely hindered in the brains of prion disease, which may contribute, at least partially, to the neuron death.
Collapse
Affiliation(s)
- Ting-Ting Wang
- School of Basic Medical Sciences, Inner Mongolia Medical University, Jinshan Development Zone, Hohhot 010110, People's Republic of China; State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, People's Republic of China
| | - Chan Tian
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, People's Republic of China; Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, People's Republic of China
| | - Jing Sun
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, People's Republic of China
| | - Hui Wang
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, People's Republic of China
| | - Bao-Yun Zhang
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, People's Republic of China
| | - Cao Chen
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, People's Republic of China
| | - Jing Wang
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, People's Republic of China
| | - Kang Xiao
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, People's Republic of China
| | - Li-Na Chen
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, People's Republic of China
| | - Yan Lv
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, People's Republic of China
| | - Chen Gao
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, People's Republic of China
| | - Qi Shi
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, People's Republic of China
| | - Yan Xin
- School of Basic Medical Sciences, Inner Mongolia Medical University, Jinshan Development Zone, Hohhot 010110, People's Republic of China.
| | - Xiao-Ping Dong
- State Key Laboratory for Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases (Zhejiang University), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 155 Changbai Rd, Beijing 102206, People's Republic of China; Chinese Academy of Sciences Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China.
| |
Collapse
|
10
|
DiFazio M, Silverberg ND, Kirkwood MW, Bernier R, Iverson GL. Prolonged Activity Restriction After Concussion: Are We Worsening Outcomes? Clin Pediatr (Phila) 2016; 55:443-51. [PMID: 26130391 DOI: 10.1177/0009922815589914] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The current treatment of concussion or mild traumatic brain injury (mTBI) is primarily based on expert consensus. Most clinical practice guidelines advise cognitive and physical rest after injury including withdrawal from normal life activities such as school attendance, sports participation, and technology use until symptoms resolve. Some individuals who sustain an mTBI experience persistent physical, cognitive, and mental health problems. Activity restriction itself may contribute to protracted recovery and other complications. Williamson's Activity Restriction Model of Depression, formulated more than 20 years ago, is central to this hypothesis. We review research evidence for potential harms of prolonged activity restriction and report an mTBI case as an example of how an "activity restriction cascade" can unfold. According to this model, psychological consequences of removal from validating life activities, combined with physical deconditioning, contribute to the development and persistence of postconcussive symptoms after mTBI in some youth. A modification to mTBI guidelines that emphasizes prompt reengagement in life activities as tolerated is encouraged.
Collapse
Affiliation(s)
- Marc DiFazio
- Children's National Health System, Washington, DC, USA
| | - Noah D Silverberg
- University of British Columbia, Vancouver, British Columbia, Canada GF Strong Rehab Centre, Vancouver, British Columbia, Canada
| | - Michael W Kirkwood
- University of Colorado School of Medicine, Aurora, CO, USA Children's Hospital Colorado, Aurora, CO, USA
| | | | - Grant L Iverson
- Harvard Medical School, Boston, MA, USA Spaulding Rehabilitation Hospital, Boston, MA, USA MassGeneral Hospital for Children, Boston, MA, USA Defense and Veterans Brain Injury Center, Bethesda, MD, USA
| |
Collapse
|
11
|
Mesenchymal stem cells secrete brain-derived neurotrophic factor and promote retinal ganglion cell survival after traumatic optic neuropathy. J Craniofac Surg 2015; 26:548-52. [PMID: 25723663 DOI: 10.1097/scs.0000000000001348] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVE The study aimed to investigate whether intravitreal injection of mesenchymal stem cells (MSCs) cultivated in vitro could increase the number of survived retinal ganglion cells (RGCs) after traumatic optic neuropathy and sought to identify potential mechanisms underlying such growth. METHODS The right eye of 24 cats in the MSC transplantation group accepted intravitreal injection of MSCs, and the other 24 cats in the phosphate buffer (PBS) control group received isotonic saline after traumatic optic neuropathy. The RGCs' survival rate in separated retinal and brain-derived neurotrophic factor (BDNF) expression were observed by Dil labeling and Enzyme-Linked Immuno Sorbent Assay (ELISA), respectively, at 3, 7, 14, and 28 days after transplantation. RESULTS Quantitative analysis showed that RGCs were significantly attenuated at 3, 7, 14, and 28 days after transplantation in both areas of the retina (peripheral, P7d = 0.0011, P14d < 0.001, P28d < 0.001; central, P3d = 0.0437, P7d = 0.0067, P14d < 0.001, P28d < 0.001). Mean density of RGCs in the MSC transplantation group was significantly higher than that of the PBS control group after 14 days of treatment (P < 0.001). The homogenates BDNF (hBDNF) in the MSC transplantation group was obviously higher than that in the PBS control group at 14 and 28 days (P < 0.05). CONCLUSIONS The MSCs transplanted into the retina of cats can slow down RGC apoptosis and steadily express BDNF. The MSC-mediated neuroprotection after optic nerve injury may be related to BNDF.
Collapse
|
12
|
Simon D, Nascimento RIMD, Filho EMR, Bencke J, Regner A. Plasma brain-derived neurotrophic factor levels after severe traumatic brain injury. Brain Inj 2015; 30:23-8. [PMID: 26555864 DOI: 10.3109/02699052.2015.1077993] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Severe traumatic brain injury (TBI) is associated with a 30-70% mortality rate. Nevertheless, in clinical practice there are no effective biomarkers for the prediction of fatal outcome following severe TBI. Therefore, the aim was to determine whether brain-derived neurotrophic factor (BDNF) plasma levels are associated with intensive care unit (ICU) mortality in patients with severe TBI. METHODS This prospective study enrolled 120 male patients who suffered severe TBI (Glasgow Coma Scale 3-8 at emergency room admission). The plasma BDNF level was determined at ICU admission (mean 6.4 hours after emergency room admission). RESULTS Severe TBI was associated with a 35% mortality rate and 64% of the patients presented severe TBI with multi-trauma. The mean plasma BDNF concentration among the severe TBI victims was 704.2 ± 63.4 pg ml(-1) (±SEM). Nevertheless, there were no significant differences between BDNF levels in the survivor (700.2 ± 82.8 pg ml(-1)) or non-survivor (711.6 ± 97.4 pg ml(-1)) groups (p = 0.238) or in the isolated TBI (800.4 ± 117.4 pg ml(-1)) or TBI with multi-trauma groups (650.5 ± 73.9 pg ml(-1)) (p = 0.109). CONCLUSIONS Plasma BDNF concentrations did not correlate with either short-term fatal outcome or type of injury following severe TBI.
Collapse
Affiliation(s)
- Daniel Simon
- a Programa de Pós-Graduação em Biologia Celular e Molecular Aplicada à Saúde .,b Laboratório de Biomarcadores do Trauma , Universidade Luterana do Brasil , Canoas , Brazil , and.,c Curso de Medicina , Universidade Luterana do Brasil , Canoas , Brazil
| | | | | | - Jane Bencke
- b Laboratório de Biomarcadores do Trauma , Universidade Luterana do Brasil , Canoas , Brazil , and
| | - Andrea Regner
- a Programa de Pós-Graduação em Biologia Celular e Molecular Aplicada à Saúde .,b Laboratório de Biomarcadores do Trauma , Universidade Luterana do Brasil , Canoas , Brazil , and.,c Curso de Medicina , Universidade Luterana do Brasil , Canoas , Brazil
| |
Collapse
|
13
|
Gram MG, Wogensen E, Wörtwein G, Mogensen J, Malá H. Delayed restraint procedure enhances cognitive recovery of spatial function after fimbria-fornix transection. Restor Neurol Neurosci 2015; 34:1-17. [PMID: 26518669 DOI: 10.3233/rnn-140396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE To i) evaluate the effect of a restraint procedure (7 days, 2 h/day) on place learning after fimbria-fornix transection (FF), ii) investigate effects of early vs. late administration of restraint, and iii) establish effects of the restraint procedure on expression of brain derived neurotrophic factor (BDNF) in prefrontal cortex and hippocampus. METHODS Fifty rats subjected to FF or sham surgery and divided into groups exposed to restraint immediately (early restraint) or 21 days (late restraint) after surgery were trained to acquire an allocentric place learning task. In parallel, 29 animals were subjected to FF or sham surgery and an identical restraint procedure in order to measure concentrations of BDNF and corticosterone. RESULTS The performance of the sham operated rats was positively affected by the late restraint. In FF-lesioned animals, the late restraint significantly improved task performance compared to the lesioned group with no restraint, while the early restraint was associated with a negative impact on task acquisition. Biochemical analysis after restraint procedure revealed a lesion-induced upregulation of BDNF in FF animals. CONCLUSIONS The improved task performance of lesioned animals suggests a therapeutic effect of this manipulation, independent of BDNF. This effect is sensitive to the temporal administration of treatment.
Collapse
Affiliation(s)
- Marie Gajhede Gram
- The Unit for Cognitive Neuroscience, Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | - Elise Wogensen
- The Unit for Cognitive Neuroscience, Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | - Gitta Wörtwein
- Laboratory of Neuropsychiatry, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Mogensen
- The Unit for Cognitive Neuroscience, Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | - Hana Malá
- The Unit for Cognitive Neuroscience, Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
14
|
Larson-Dupuis C, Chamard É, Falardeau V, Frasnelli J, Beaulieu C, Poirier J, Carrier J, Lassonde M, Théoret H, Bacon BA, De Beaumont L. Impact of BDNF Val66Met polymorphism on olfactory functions of female concussed athletes. Brain Inj 2015; 29:963-70. [DOI: 10.3109/02699052.2015.1016452] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
15
|
Gagnon I, Grilli L, Friedman D, Iverson GL. A pilot study of active rehabilitation for adolescents who are slow to recover from sport-related concussion. Scand J Med Sci Sports 2015; 26:299-306. [DOI: 10.1111/sms.12441] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2015] [Indexed: 11/28/2022]
Affiliation(s)
- I. Gagnon
- School of Physical and Occupational Therapy; Faculty of Medicine; McGill University; Montreal Quebec Canada
- Trauma Center; The Montreal Children's Hospital; McGill University Health Center; Montreal Quebec Canada
- Department of Pediatrics; Faculty of Medicine; McGill University; Montreal Quebec Canada
| | - L. Grilli
- Trauma Center; The Montreal Children's Hospital; McGill University Health Center; Montreal Quebec Canada
| | - D. Friedman
- Trauma Center; The Montreal Children's Hospital; McGill University Health Center; Montreal Quebec Canada
- Department of Pediatrics; Faculty of Medicine; McGill University; Montreal Quebec Canada
- Canadian Hospitals Injury Reporting and Prevention Program; Montreal Quebec Canada
| | - G. L. Iverson
- Department of Physical Medicine and Rehabilitation; Harvard Medical School; Boston Massachusetts USA
- Spaulding Rehabilitation Hospital; Boston Massachusetts USA
- Mass General Hospital for Children Sports Concussion Program; Boston Massachusetts USA
- Red Sox Foundation and Massachusetts General Hospital Home Base Program; Boston Massachusetts USA
| |
Collapse
|
16
|
Pinelis VG, Sorokina EG, Semenova JB, Karaseva OV, Mescheryakov SV, Chernisheva TA, Arsenieva EN, Roshal LM. Biomarkers in children with traumatic brain injury. Zh Nevrol Psikhiatr Im S S Korsakova 2015; 115:66-72. [DOI: 10.17116/jnevro20151158166-72] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
17
|
Moser RS, Schatz P, Glenn M, Kollias KE, Iverson GL. Examining prescribed rest as treatment for adolescents who are slow to recover from concussion. Brain Inj 2014; 29:58-63. [DOI: 10.3109/02699052.2014.964771] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
| | | | - Megan Glenn
- Sports Concussion Center of New Jersey, RSM Psychology Center, LLC, Lawrenceville, NJ, USA,
| | - Kelly E. Kollias
- Sports Concussion Center of New Jersey, RSM Psychology Center, LLC, Lawrenceville, NJ, USA,
| | - Grant L. Iverson
- Department of Physical Medicine and Rehabilitation, Harvard Medical School, Harvard, Boston, MA, USA,
- Spaulding Rehabilitation Hospital,
- Massachusetts General Hospital Sport Concussion Clinic, Boston, MA, USA, and
- Red Sox Foundation and Massachusetts General Hospital Home Base Program, Boston, MA, USA
| |
Collapse
|
18
|
Zhang H, Han M, Zhang X, Sun X, Ling F. The effect and mechanism of growth hormone replacement on cognitive function in rats with traumatic brain injury. PLoS One 2014; 9:e108518. [PMID: 25268832 PMCID: PMC4182486 DOI: 10.1371/journal.pone.0108518] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 08/31/2014] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE The effects of growth hormone on cognitive dysfunction were observed in a controlled cortical impact (CCI) rat model and the underlying mechanism was explored. METHOD Three-month-old male SD rats were randomly divided into sham (n = 10), control (n = 10), and CCI groups (n = 40) The parameters were set as follows: striking speed, 3.5 m/s; impact depth, 1.5 mm; and dwell time, 400 msec. Eight and ten weeks post-injury, the GH levels were measured the water maze test and novel object recognition test were performed. CCI rats were divided into normal and decreased GH groups, and further randomly divided into two sub-groups (rhGH treatment and saline vehicle groups). All rats were tested for SYN, BDNF, and TrkB mRNA in the prefrontal cortex and hippocampus by RT-PCR. RESULTS CCI rats 8 weeks post-injury had cognitive dysfunction regardless of the GH level (P<0.05). rhGH treatment improved cognitive function in CCI rats. There was a positive correlation between the expression of prefrontal BDNF and SYN mRNA in CCI rats after rhGH therapy and the water maze test score (r = 0.773 and 0.534, respectively; P<0.05). Furthermore, the expression of BDNF, TrkB, and SYN mRNA in the hippocampus was negatively correlated with the water maze test score (r = 0.602, 0.773, 0.672, and 0.783, respectively; P<0.05). There was a difference in the expression of hippocampal and prefrontal BDNF, TrkB, and SYN mRNA (P<0.05). CONCLUSION rhGH treatment had a positive effect on cognitive function, which was more evident in GH-deficient rats. The increased expression of hippocampal and prefrontal BDNF and TrkB mRNA is implicated in rhGH therapy to improve cognitive function. Changes in the expression of hippocampal SYN mRNA following rhGH therapy may also play a role in improving cognitive function.
Collapse
Affiliation(s)
- Hao Zhang
- China Rehabilitation Research Center, Capital Medical University, Beijing, China
| | - Mengqi Han
- Beijing Jishuitan Hospital, Beijing, China
| | - Xiaonian Zhang
- China Rehabilitation Research Center, Capital Medical University, Beijing, China
| | - Xinting Sun
- China Rehabilitation Research Center, Capital Medical University, Beijing, China
| | - Feng Ling
- Beijing Xuanwu Hospital, Capital Medical University, Beijing, China
- * E-mail:
| |
Collapse
|
19
|
Chan JL, Reeves TM, Phillips LL. Osteopontin expression in acute immune response mediates hippocampal synaptogenesis and adaptive outcome following cortical brain injury. Exp Neurol 2014; 261:757-71. [PMID: 25151457 DOI: 10.1016/j.expneurol.2014.08.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 08/04/2014] [Accepted: 08/14/2014] [Indexed: 01/13/2023]
Abstract
Traumatic brain injury (TBI) produces axotomy, deafferentation and reactive synaptogenesis. Inflammation influences synaptic repair, and the novel brain cytokine osteopontin (OPN) has potential to support axon regeneration through exposure of its integrin receptor binding sites. This study explored whether OPN secretion and proteolysis by matrix metalloproteinases (MMPs) mediate the initial degenerative phase of synaptogenesis, targeting reactive neuroglia to affect successful repair. Adult rats received unilateral entorhinal cortex lesion (UEC) modeling adaptive synaptic plasticity. Over the first week postinjury, hippocampal OPN protein and mRNA were assayed and histology was performed. At 1-2d, OPN protein increased up to 51 fold, and was localized within activated, mobilized glia. OPN transcript also increased over 50 fold, predominantly within reactive microglia. OPN fragments known to be derived from MMP proteolysis were elevated at 1d, consistent with prior reports of UEC glial activation and enzyme production. Postinjury minocycline immunosuppression attenuated MMP-9 gelatinase activity, which was correlated with the reduction of neutrophil gelatinase-associated lipocalin (LCN2) expression, and reduced OPN fragment generation. The antibiotic also attenuated removal of synapsin-1 positive axons from the deafferented zone. OPN KO mice subjected to UEC had similar reduction of hippocampal MMP-9 activity, as well as lower synapsin-1 breakdown over the deafferented zone. MAP1B and N-cadherin, surrogates of cytoarchitecture and synaptic adhesion, were not affected. OPN KO mice with UEC exhibited time dependent cognitive deficits during the synaptogenic phase of recovery. This study demonstrates that OPN can mediate immune response during TBI synaptic repair, positively influencing synapse reorganization and functional recovery.
Collapse
Affiliation(s)
- Julie L Chan
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, P.O. Box 980709, Richmond, VA 23298, USA
| | - Thomas M Reeves
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, P.O. Box 980709, Richmond, VA 23298, USA
| | - Linda L Phillips
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, P.O. Box 980709, Richmond, VA 23298, USA.
| |
Collapse
|
20
|
Edut S, Rubovitch V, Rehavi M, Schreiber S, Pick CG. A study on the mechanism by which MDMA protects against dopaminergic dysfunction after minimal traumatic brain injury (mTBI) in mice. J Mol Neurosci 2014; 54:684-97. [PMID: 25124230 DOI: 10.1007/s12031-014-0399-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 08/05/2014] [Indexed: 12/11/2022]
Abstract
Driving under methylenedioxymethamphetamine (MDMA) influence increases the risk of being involved in a car accident, which in turn can lead to traumatic brain injury. The behavioral deficits after traumatic brain injury (TBI) are closely connected to dopamine pathway dysregulation. We have previously demonstrated in mice that low MDMA doses prior to mTBI can lead to better performances in cognitive tests. The purpose of this study was to assess in mice the changes in the dopamine system that occurs after both MDMA and minimal traumatic brain injury (mTBI). Experimental mTBI was induced using a concussive head trauma device. One hour before injury, animals were subjected to MDMA. Administration of MDMA before injury normalized the alterations in tyrosine hydroxylase (TH) levels that were observed in mTBI mice. This normalization was also able to lower the elevated dopamine receptor type 2 (D2) levels observed after mTBI. Brain-derived neurotrophic factor (BDNF) levels did not change following injury alone, but in mice subjected to MDMA and mTBI, significant elevations were observed. In the behavioral tests, haloperidol reversed the neuroprotection seen when MDMA was administered prior to injury. Altered catecholamine synthesis and high D2 receptor levels contribute to cognitive dysfunction, and strategies to normalize TH signaling and D2 levels may provide relief for the deficits observed after injury. Pretreatment with MDMA kept TH and D2 receptor at normal levels, allowing regular dopamine system activity. While the beneficial effect we observe was due to a dangerous recreational drug, understanding the alterations in dopamine and the mechanism of dysfunction at a cellular level can lead to legal therapies and potential candidates for clinical use.
Collapse
Affiliation(s)
- S Edut
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel,
| | | | | | | | | |
Collapse
|
21
|
Macedo RCD, Tomasi CD, Giombelli VR, Alves SC, Bristot MDLU, Locks MFT, Petronilho F, Grandi C, Quevedo J, Dal-Pizzol F, Ritter C. Lack of association of S100β and neuron-specific enolase with mortality in critically ill patients. BRAZILIAN JOURNAL OF PSYCHIATRY 2014; 35:267-70. [PMID: 24142088 DOI: 10.1590/1516-4446-2012-0835] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 05/15/2012] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To evaluate the relationship between brain damage biomarkers and mortality in the intensive care unit (ICU). METHODS The sample comprised 70 patients admitted to an ICU. Blood samples were collected from all patients on ICU admission, and levels of S100β and neuron-specific enolase (NSE) were determined by ELISA. RESULTS Acute Physiologic and Chronic Health Evaluation (APACHE II) score was associated with mortality, but NSE and S100β were not associated with this outcome. In contrast, S100β levels were significantly higher in delirious and non-delirious patients who required mechanical ventilation during ICU stay. CONCLUSION Levels of brain biomarkers at the time of ICU admission did not predict mortality in critically ill patients.
Collapse
Affiliation(s)
- Roberta Candal de Macedo
- National Science and Technology Institute for Translational Medicine, Laboratory of Experimental Pathophysiology, CriciúmaSC, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
BDNF mimetic compound LM22A-4 regulates cementoblast differentiation via the TrkB–ERK/Akt signaling cascade. Int Immunopharmacol 2014; 19:245-52. [DOI: 10.1016/j.intimp.2014.01.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 01/11/2014] [Accepted: 01/30/2014] [Indexed: 11/21/2022]
|
23
|
Shen X, Li A, Zhang Y, Dong X, Shan T, Wu Y, Jia J, Hu Y. The effect of different intensities of treadmill exercise on cognitive function deficit following a severe controlled cortical impact in rats. Int J Mol Sci 2013; 14:21598-612. [PMID: 24185909 PMCID: PMC3856023 DOI: 10.3390/ijms141121598] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 10/12/2013] [Accepted: 10/17/2013] [Indexed: 11/16/2022] Open
Abstract
Exercise has been proposed for the treatment of traumatic brain injury (TBI). However, the proper intensity of exercise in the early phase following a severe TBI is largely unknown. To compare two different treadmill exercise intensities on the cognitive function following a severe TBI in its early phase, rats experienced a controlled cortical impact (CCI) and were forced to treadmill exercise for 14 days. The results revealed that the rats in the low intensity exercise group had a shorter latency to locate a platform and a significantly better improvement in spatial memory in the Morris water maze (MWM) compared to the control group (p < 0.05). The high intensity exercise group showed a longer latency and a mild improvement in spatial memory compared to the control group rats in the MWM; however, this difference was not statistically significant (p > 0.05). The brain-derived neurotrophic factor (BDNF) and p-CREB protein levels in the contralateral hippocampus were increased significantly in the low intensity exercise group. Our results suggest that 2 weeks of low intensity of treadmill exercise is beneficial for improving cognitive function and increasing hippocampal BDNF expression after a severe TBI in its early phase.
Collapse
Affiliation(s)
- Xiafeng Shen
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mails: (X.S.); (Y.Z.); (T.S.); (Y.W.); (J.J.); (Y.H.)
- Department of Rehabilitation, Shanghai Yangpu District Geriatric Hospital, Shanghai 200090, China
| | - Aiping Li
- Rehabilitation Medicine Center, Nanjing Military Region Sanatorium of Hangzhou, Hangzhou 310007, Zhejiang, China; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +86-571-8734-8142; Fax: +86-571-8734-8114
| | - Yuling Zhang
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mails: (X.S.); (Y.Z.); (T.S.); (Y.W.); (J.J.); (Y.H.)
| | - XiaoMin Dong
- Rehabilitation Medicine Center, Nanjing Military Region Sanatorium of Hangzhou, Hangzhou 310007, Zhejiang, China; E-Mail:
| | - Tian Shan
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mails: (X.S.); (Y.Z.); (T.S.); (Y.W.); (J.J.); (Y.H.)
| | - Yi Wu
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mails: (X.S.); (Y.Z.); (T.S.); (Y.W.); (J.J.); (Y.H.)
| | - Jie Jia
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mails: (X.S.); (Y.Z.); (T.S.); (Y.W.); (J.J.); (Y.H.)
| | - Yongshan Hu
- Department of Rehabilitation, Huashan Hospital, Fudan University, Shanghai 200040, China; E-Mails: (X.S.); (Y.Z.); (T.S.); (Y.W.); (J.J.); (Y.H.)
| |
Collapse
|
24
|
Budinich CS, Tucker LB, Lowe D, Rosenberger JG, McCabe JT. Short and long-term motor and behavioral effects of diazoxide and dimethyl sulfoxide administration in the mouse after traumatic brain injury. Pharmacol Biochem Behav 2013; 108:66-73. [DOI: 10.1016/j.pbb.2013.04.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 04/02/2013] [Accepted: 04/06/2013] [Indexed: 01/11/2023]
|
25
|
TrkB receptor signalling: implications in neurodegenerative, psychiatric and proliferative disorders. Int J Mol Sci 2013; 14:10122-42. [PMID: 23670594 PMCID: PMC3676832 DOI: 10.3390/ijms140510122] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 04/27/2013] [Accepted: 04/28/2013] [Indexed: 02/06/2023] Open
Abstract
The Trk family of receptors play a wide variety of roles in physiological and disease processes in both neuronal and non-neuronal tissues. Amongst these the TrkB receptor in particular has attracted major attention due to its critical role in signalling for brain derived neurotrophic factor (BDNF), neurotrophin-3 (NT3) and neurotrophin-4 (NT4). TrkB signalling is indispensable for the survival, development and synaptic plasticity of several subtypes of neurons in the nervous system. Substantial evidence has emerged over the last decade about the involvement of aberrant TrkB signalling and its compromise in various neuropsychiatric and degenerative conditions. Unusual changes in TrkB signalling pathway have also been observed and implicated in a range of cancers. Variations in TrkB pathway have been observed in obesity and hyperphagia related disorders as well. Both BDNF and TrkB have been shown to play critical roles in the survival of retinal ganglion cells in the retina. The ability to specifically modulate TrkB signalling can be critical in various pathological scenarios associated with this pathway. In this review, we discuss the mechanisms underlying TrkB signalling, disease implications and explore plausible ameliorative or preventive approaches.
Collapse
|
26
|
Álvarez XA, Figueroa J, Muresanu D. Peptidergic drugs for the treatment of traumatic brain injury. FUTURE NEUROLOGY 2013. [DOI: 10.2217/fnl.12.95] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Traumatic brain injury (TBI) is a devastating medical condition that has an enormous socioeconomic impact because it affects more than 10 million people annually worldwide and is associated with high rates of hospitalization, mortality and disability. Although TBI survival has improved continuously for decades, particularly in developing countries, implementation of an effective drug therapy for TBI represents an unmet clinical need. All confirmatory trials conducted to date with drugs targeting a single TBI pathological pathway failed to show clinical efficacy, probably because TBI pathophysiology involves multiple cellular and molecular mechanisms of secondary brain damage. According to current scientific evidence of the participation of peptide-mediated mechanisms in the processes of brain injury and repair after TBI, peptidergic drugs represent a multimodal therapy alternative to improve acute outcome and long-term recovery in TBI patients. Preliminary randomized-controlled clinical trials and open-label studies conducted to date with the peptidergic drug Cerebrolysin® (Ever Neuro Pharma GmbH, Unterach, Austria) and with the endogenous neuropeptides progesterone and erythropoietin, showed positive clinical results. Cerebrolysin-treated patients had a faster clinical recovery, a shorter hospitalization time and a better long-term outcome. Treatment with progesterone showed advantages over placebo regarding TBI mortality and clinical outcome, whereas erythropoietin only reduced mortality. Further validation of these promising findings in confirmatory randomized-controlled clinical trials is warranted. This article reviews the scientific basis and clinical evidence on the development of multimodal peptidergic drugs as a therapeutic option for the effective treatment of TBI patients.
Collapse
Affiliation(s)
| | - Jesús Figueroa
- Rehabilitation Department, Santiago University Hospital, Santiago de Compostela, Spain
| | - Dafin Muresanu
- Department of Neurology, University of Medicine & Pharmacy ‘Iuliu Hatieganu’, Cluj-Napoca, Romania
| |
Collapse
|
27
|
Schober ME, Block B, Requena DF, Hale MA, Lane RH. Developmental traumatic brain injury decreased brain derived neurotrophic factor expression late after injury. Metab Brain Dis 2012; 27:167-73. [PMID: 22527999 PMCID: PMC3383795 DOI: 10.1007/s11011-012-9309-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Accepted: 04/15/2012] [Indexed: 01/06/2023]
Abstract
Pediatric traumatic brain injury (TBI) is a major cause of acquired cognitive dysfunction in children. Hippocampal Brain Derived Neurotrophic Factor (BDNF) is important for normal cognition. Little is known about the effects of TBI on BDNF levels in the developing hippocampus. We used controlled cortical impact (CCI) in the 17 day old rat pup to test the hypothesis that CCI would first increase rat hippocampal BDNF mRNA/protein levels relative to SHAM and Naïve rats by post injury day (PID) 2 and then decrease BDNF mRNA/protein by PID14. Relative to SHAM, CCI did not change BDNF mRNA/protein levels in the injured hippocampus in the first 2 days after injury but did decrease BDNF protein at PID14. Surprisingly, BDNF mRNA decreased at PID 1, 3, 7 and 14, and BDNF protein decreased at PID 2, in SHAM and CCI hippocampi relative to Naïve. In conclusion, TBI decreased BDNF protein in the injured rat pup hippocampus 14 days after injury. BDNF mRNA levels decreased in both CCI and SHAM hippocampi relative to Naïve, suggesting that certain aspects of the experimental paradigm (such as craniotomy, anesthesia, and/or maternal separation) may decrease the expression of BDNF in the developing hippocampus. While BDNF is important for normal cognition, no inferences can be made regarding the cognitive impact of any of these factors. Such findings, however, suggest that meticulous attention to the experimental paradigm, and possible inclusion of a Naïve group, is warranted in studies of BDNF expression in the developing brain after TBI.
Collapse
|
28
|
Neuroprotection and neuroregeneration: what to expect from a stem cell-based therapy of acute brain injury. Crit Care Med 2011; 39:2577-8. [PMID: 22005237 DOI: 10.1097/ccm.0b013e31822e56f3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
29
|
Zanier ER, Montinaro M, Vigano M, Villa P, Fumagalli S, Pischiutta F, Longhi L, Leoni ML, Rebulla P, Stocchetti N, Lazzari L, De Simoni MG. Human umbilical cord blood mesenchymal stem cells protect mice brain after trauma. Crit Care Med 2011; 39:2501-10. [PMID: 21725237 DOI: 10.1097/ccm.0b013e31822629ba] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To investigate whether human umbilical cord blood mesenchymal stem cells, a novel source of progenitors with multilineage potential: 1) decrease traumatic brain injury sequelae and restore brain function; 2) are able to survive and home to the lesioned region; and 3) induce relevant changes in the environment in which they are infused. DESIGN Prospective experimental study. SETTING Research laboratory. SUBJECTS Male C57Bl/6 mice. INTERVENTIONS Mice were subjected to controlled cortical impact/sham brain injury. At 24 hrs postinjury, human umbilical cord blood mesenchymal stem cells (150,000/5 μL) or phosphate-buffered saline (control group) were infused intracerebroventricularly contralateral to the injured side. Immunosuppression was achieved by cyclosporine A (10 mg/kg intraperitoneally). MEASUREMENTS AND MAIN RESULTS After controlled cortical impact, human umbilical cord blood mesenchymal stem cell transplantation induced an early and long-lasting improvement in sensorimotor functions assessed by neuroscore and beam walk tests. One month postinjury, human umbilical cord blood mesenchymal stem cell mice showed attenuated learning dysfunction at the Morris water maze and reduced contusion volume compared with controls. Hoechst positive human umbilical cord blood mesenchymal stem cells homed to lesioned tissue as early as 1 wk after injury in 67% of mice and survived in the injured brain up to 5 wks. By 3 days postinjury, cell infusion significantly increased brain-derived neurotrophic factor concentration into the lesioned tissue, restoring its expression close to the levels observed in sham operated mice. By 7 days postinjury, controlled cortical impact human umbilical cord blood mesenchymal stem cell mice showed a nonphagocytic activation of microglia/macrophages as shown by a selective rise (260%) in CD11b staining (a marker of microglia/macrophage activation/recruitment) associated with a decrease (58%) in CD68 (a marker of active phagocytosis). Thirty-five days postinjury, controlled cortical impact human umbilical cord blood mesenchymal stem cell mice showed a decrease of glial fibrillary acidic protein positivity in the scar region compared with control mice. CONCLUSIONS These findings indicate that human umbilical cord blood mesenchymal stem cells stimulate the injured brain and evoke trophic events, microglia/macrophage phenotypical switch, and glial scar inhibitory effects that remodel the brain and lead to significant improvement of neurologic outcome.
Collapse
Affiliation(s)
- Elisa R Zanier
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milano, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Albert-Weissenberger C, Sirén AL. Experimental traumatic brain injury. EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2010; 2:16. [PMID: 20707892 PMCID: PMC2930598 DOI: 10.1186/2040-7378-2-16] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Accepted: 08/13/2010] [Indexed: 12/03/2022]
Abstract
Traumatic brain injury, a leading cause of death and disability, is a result of an outside force causing mechanical disruption of brain tissue and delayed pathogenic events which collectively exacerbate the injury. These pathogenic injury processes are poorly understood and accordingly no effective neuroprotective treatment is available so far. Experimental models are essential for further clarification of the highly complex pathology of traumatic brain injury towards the development of novel treatments. Among the rodent models of traumatic brain injury the most commonly used are the weight-drop, the fluid percussion, and the cortical contusion injury models. As the entire spectrum of events that might occur in traumatic brain injury cannot be covered by one single rodent model, the design and choice of a specific model represents a major challenge for neuroscientists. This review summarizes and evaluates the strengths and weaknesses of the currently available rodent models for traumatic brain injury.
Collapse
|
31
|
|
32
|
Massa SM, Yang T, Xie Y, Shi J, Bilgen M, Joyce JN, Nehama D, Rajadas J, Longo FM. Small molecule BDNF mimetics activate TrkB signaling and prevent neuronal degeneration in rodents. J Clin Invest 2010; 120:1774-85. [PMID: 20407211 DOI: 10.1172/jci41356] [Citation(s) in RCA: 308] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Accepted: 02/17/2010] [Indexed: 02/01/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) activates the receptor tropomyosin-related kinase B (TrkB) with high potency and specificity, promoting neuronal survival, differentiation, and synaptic function. Correlations between altered BDNF expression and/or function and mechanism(s) underlying numerous neurodegenerative conditions, including Alzheimer disease and traumatic brain injury, suggest that TrkB agonists might have therapeutic potential. Using in silico screening with a BDNF loop-domain pharmacophore, followed by low-throughput in vitro screening in mouse fetal hippocampal neurons, we have efficiently identified small molecules with nanomolar neurotrophic activity specific to TrkB versus other Trk family members. Neurotrophic activity was dependent on TrkB and its downstream targets, although compound-induced signaling activation kinetics differed from those triggered by BDNF. A selected prototype compound demonstrated binding specificity to the extracellular domain of TrkB. In in vitro models of neurodegenerative disease, it prevented neuronal degeneration with efficacy equal to that of BDNF, and when administered in vivo, it caused hippocampal and striatal TrkB activation in mice and improved motor learning after traumatic brain injury in rats. These studies demonstrate the utility of loop modeling in drug discovery and reveal what we believe to be the first reported small molecules derived from a targeted BDNF domain that specifically activate TrkB.We propose that these compounds constitute a novel group of tools for the study of TrkB signaling and may provide leads for developing new therapeutic agents for neurodegenerative diseases.
Collapse
Affiliation(s)
- Stephen M Massa
- Department of Neurology and Laboratory for Computational Neurochemistry and Drug Discovery, UCSF, San Francisco, California, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Kalish H, Phillips TM. Analysis of neurotrophins in human serum by immunoaffinity capillary electrophoresis (ICE) following traumatic head injury. J Chromatogr B Analyt Technol Biomed Life Sci 2009; 878:194-200. [PMID: 19896422 DOI: 10.1016/j.jchromb.2009.10.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Revised: 10/20/2009] [Accepted: 10/21/2009] [Indexed: 10/20/2022]
Abstract
Neurotrophins, including brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF), neurotrophin-3 (NT-3), neurotrophin-4 (NT-4), and beta-nerve growth factor (beta-NGF), play an active role in the development, maintenance and survival of cells of the central nervous system (CNS). Previous research has indicated that a decrease in concentrations of these neurotrophins is often associated with cell death and ultimately patient demise. However, much of the research conducted analyses of samples taken directly from the CNS, i.e., of samples that are not readily available in clinical trauma centers. In an attempt to obtain a method for evaluating neurotrophins in a more readily accessible matrix, i.e., serum, a precise and accurate immunoaffinity capillary electrophoresis (ICE) method was developed and applied to measure neurotrophins in serum from patients with various degrees of head injury. The five neurotrophins of interest were extracted and concentrated by specific immunochemically immobilized antibodies, bound directly to the capillary wall, and eluted and separated in approximately 10min. NT-3, BDNF, CNTF and beta-NGF showed a marked decrease in concentration as the severity of the head injury increased: mild versus severe: 91% decrease for NT-3; 93 % decrease for BDNF; 93 % decrease for CNTF; and a 87 % decrease for beta-NGF. This decrease in concentration is consistent with the neuro-protective roles that neurotrophins play in the maintenance and survival of neuronal cells. The results obtained by the ICE method were closely comparable with those generated by a commercially available ELISA method.
Collapse
Affiliation(s)
- Heather Kalish
- Ultramicro Immunodiagnostics Section, Laboratory of Bioengineering & Physical Science, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD USA.
| | | |
Collapse
|
34
|
Griesbach GS, Hovda DA, Gomez-Pinilla F. Exercise-induced improvement in cognitive performance after traumatic brain injury in rats is dependent on BDNF activation. Brain Res 2009; 1288:105-15. [PMID: 19555673 DOI: 10.1016/j.brainres.2009.06.045] [Citation(s) in RCA: 207] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2009] [Revised: 06/09/2009] [Accepted: 06/11/2009] [Indexed: 10/20/2022]
Abstract
We have previously shown that voluntary exercise upregulates brain derived neurotrophic factor (BDNF) within the hippocampus and is associated with an enhancement of cognitive recovery after a lateral fluid percussion injury (FPI). In order to determine if BDNF is critical to this effect we used an immunoadhesin chimera (TrkB-IgG) that inactivates free BDNF. This BDNF inhibitor was administered to adult male rats two weeks after they had received a mild fluid percussion injury (FPI) or sham surgery. These animals were then housed with or without access to a running wheel (RW) from post-injury-day (PID) 14 to 20. On PID 21, rats were tested for spatial learning in a Morris Water Maze. Results showed that exercise counteracted the cognitive deficits associated with the injury. However this exercise-induced cognitive improvement was attenuated in the FPI-RW rats that were treated with TrkB-IgG. Molecules important for synaptic plasticity and learning were measured in a separate group of rats that were sacrificed immediately after exercise (PID 21). Western blot analyses showed that exercise increased the mature form of BDNF, synapsin I and cyclic-AMP response-element-binding protein (CREB) in the vehicle treated Sham-RW group. However, only the mature form of BDNF and CREB were increased in the vehicle treated FPI-RW group. Blocking BDNF (pre administration of TrkB-IgG) greatly reduced the molecular effects of exercise in that exercise-induced increases of BDNF, synapsin I and CREB were not observed. These studies provide evidence that BDNF has a major role in exercise's cognitive effects in traumatically injured brain.
Collapse
Affiliation(s)
- Grace Sophia Griesbach
- Department of Neurosurgery, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095-7039, USA.
| | | | | |
Collapse
|