1
|
Sahbaie P, Shi X, Guo TZ, Li WW, Irvine KA, Chen Q, Clark JD. Exercise plasma improves traumatic brain injury outcomes in mice. Sci Rep 2025; 15:17957. [PMID: 40410204 PMCID: PMC12102241 DOI: 10.1038/s41598-025-02509-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 05/13/2025] [Indexed: 05/25/2025] Open
Abstract
Traumatic brain injury (TBI) is associated with pain and cognitive impairment although exercise may improve some adverse outcomes. We hypothesized that transfer of plasma derived from exercised mice would improve outcomes related to pain and memory after TBI. These studies used a concussive murine model of mild TBI. Plasma from sedentary or exercised mice was administered to TBI mice beginning 3 days after injuries. Mechanical nociceptive sensitization and descending control of nociception (DCN) were assessed. Object recognition memory and Y-maze were used to assess working memory. In addition, the spinal expression of Pdyn, Bdnf, Cxcl1 and Cxcl2 was measured 7 and 28 days after injuries. Levels of several candidate mediators of the exercise effects were measured in the transferred plasma. Mechanical allodynia following TBI recovered more quickly and DCN responses were partially preserved in animals receiving exercise plasma. Working memory was preserved in the same mice. The upregulation of spinal pain genes Pdyn and Bdnf was reduced by exercise plasma. Finally, exercise was associated with substantially higher plasma levels of TIMP2 and irisin. The present study suggests that developing therapies based on the administration of exercise plasma or its components may help reduce pain and cognitive loss after TBI.
Collapse
Affiliation(s)
- Peyman Sahbaie
- Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, 3801 Miranda Ave (112-A), Palo Alto, CA, 94304, USA.
- Palo Alto Veterans Institute for Research, 3801 Miranda Ave, Palo Alto, CA, 94304, USA.
- Department of Anesthesia, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA, 94305, USA.
| | - Xiaoyou Shi
- Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, 3801 Miranda Ave (112-A), Palo Alto, CA, 94304, USA
- Department of Anesthesia, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Tian-Zhi Guo
- Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, 3801 Miranda Ave (112-A), Palo Alto, CA, 94304, USA
- Palo Alto Veterans Institute for Research, 3801 Miranda Ave, Palo Alto, CA, 94304, USA
| | - Wen-Wu Li
- Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, 3801 Miranda Ave (112-A), Palo Alto, CA, 94304, USA
- Department of Anesthesia, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Karen-Amanda Irvine
- Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, 3801 Miranda Ave (112-A), Palo Alto, CA, 94304, USA
- Palo Alto Veterans Institute for Research, 3801 Miranda Ave, Palo Alto, CA, 94304, USA
| | - QiLiang Chen
- Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, 3801 Miranda Ave (112-A), Palo Alto, CA, 94304, USA
- Department of Anesthesia, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - J David Clark
- Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, 3801 Miranda Ave (112-A), Palo Alto, CA, 94304, USA
- Department of Anesthesia, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
2
|
Giesler LP, Mychasiuk R, Shultz SR, McDonald SJ. BDNF: New Views of an Old Player in Traumatic Brain Injury. Neuroscientist 2024; 30:560-573. [PMID: 37067029 PMCID: PMC11423547 DOI: 10.1177/10738584231164918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Traumatic brain injury is a common health problem affecting millions of people each year. BDNF has been investigated in the context of traumatic brain injury due to its crucial role in maintaining brain homeostasis. Val66Met is a functional single-nucleotide polymorphism that results in a valine-to-methionine amino acid substitution at codon 66 in the BDNF prodomain, which ultimately reduces secretion of BDNF. Here, we review experimental animal models as well as clinical studies investigating the role of the Val66Met single-nucleotide polymorphism in traumatic brain injury outcomes, including cognitive function, motor function, neuropsychiatric symptoms, and nociception. We also review studies investigating the role of BDNF on traumatic brain injury pathophysiology as well as circulating BDNF as a biomarker of traumatic brain injury.
Collapse
Affiliation(s)
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Neurology, The Alfred Hospital, Melbourne, Australia
| | - Sandy R. Shultz
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Neurology, The Alfred Hospital, Melbourne, Australia
| | - Stuart J. McDonald
- Department of Neuroscience, Monash University, Melbourne, Australia
- Department of Neurology, The Alfred Hospital, Melbourne, Australia
| |
Collapse
|
3
|
Wen J, Tanaka M, Zhang Y. Inhibition of 2-AG hydrolysis alleviates posttraumatic headache attributed to mild traumatic brain injury. J Headache Pain 2024; 25:115. [PMID: 39014318 PMCID: PMC11253377 DOI: 10.1186/s10194-024-01817-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 06/20/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Posttraumatic headache (PTH) is a common and debilitating symptom following repetitive mild traumatic brain injury (rmTBI), and it mainly resembles a migraine-like phenotype. While modulation of the endocannabinoid system (ECS) is effective in treating TBI and various types of pain including migraine, the role of augmentation of endocannabinoids in treating PTH has not been investigated. METHODS Repetitive mild TBI was induced in male C57BL/6J mice using the non-invasive close-head impact model of engineered rotational acceleration (CHIMERA). Periorbital allodynia was assessed using von Frey filaments and determined by the "Up-Down" method. Immunofluorescence staining was employed to investigate glial cell activation and calcitonin gene-related peptide (CGRP) expression in the trigeminal ganglion (TG) and trigeminal nucleus caudalis (TNC) of the rmTBI mice. Levels of 2-arachidonoyl glycerol (2-AG), anandamide (AEA), and arachidonic acid (AA) in the TG, medulla (including TNC), and periaqueductal gray (PAG) were measured by mass spectrometry. The therapeutic effect of endocannabinoid modulation on PTH was also assessed. RESULTS The rmTBI mice exhibited significantly increased cephalic pain hypersensitivity compared to the sham controls. MJN110, a potent and selective inhibitor of the 2-AG hydrolytic enzyme monoacylglycerol lipase (MAGL), dose-dependently attenuated periorbital allodynia in the rmTBI animals. Administration of CGRP at 0.01 mg/kg reinstated periorbital allodynia in the rmTBI animals on days 33 and 45 post-injury but had no effect in the sham and MJN110 treatment groups. Activation of glial cells along with increased production of CGRP in the TG and TNC at 7 and 14 days post-rmTBI were attenuated by MJN110 treatment. The anti-inflammatory and anti-nociceptive effects of MJN110 were partially mediated by cannabinoid receptor activation, and the pain-suppressive effect of MJN110 was completely blocked by co-administration of DO34, an inhibitor of 2-AG synthase. The levels of 2-AG in TG, TNC and PAG were decreased in TBI animals, significantly elevated and further reduced by the selective inhibitors of 2-AG hydrolytic and synthetic enzymes, respectively. CONCLUSION Enhancing endogenous levels of 2-AG appears to be an effective strategy for the treatment of PTH by attenuating pain initiation and transmission in the trigeminal pathway and facilitating descending pain inhibitory modulation.
Collapse
Affiliation(s)
- Jie Wen
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Mikiei Tanaka
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Yumin Zhang
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
4
|
Irvine KA, Shi XY, Ferguson AR, Clark JD. Designer Receptor Exclusively Activated by Designer Drug (DREADD)-Mediated Activation of the Periaqueductal Gray Restores Nociceptive Descending Inhibition After Traumatic Brain Injury in Rats. J Neurotrauma 2024; 41:e1761-e1779. [PMID: 38588130 PMCID: PMC11386998 DOI: 10.1089/neu.2024.0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024] Open
Abstract
Traumatic brain injury (TBI) patients frequently experience chronic pain that can enhance their suffering and significantly impair rehabilitative efforts. Clinical studies suggest that damage to the periaqueductal gray matter (PAG) following TBI, a principal center involved in endogenous pain control, may underlie the development of chronic pain. We hypothesized that TBI would diminish the usual pain control functions of the PAG, but that directly stimulating this center using a chemogenetic approach would restore descending pain modulation. We used a well-characterized lateral fluid percussion model (1.3 ± 0.1 atm) of TBI in male rats (n = 271) and measured hindpaw mechanical nociceptive withdrawal thresholds using von Frey filaments. To investigate the role of the PAG in pain both before and after TBI, we activated the neurons of the PAG using a Designer Receptor Exclusively Activated by Designer Drug (DREADD) viral construct. Immunohistochemical analysis of brain tissue was used to assess the location and confirm the appropriate expression of the viral constructs in the PAG. Activation of the PAG DREADD using clozapine N-oxide (CNO) caused hindpaw analgesia that could be blocked using opioid receptor antagonist, naloxone, in uninjured but not TBI rats. Due to the importance of descending serotonergic signaling in modulating nociception, we ablated spinal serotonin signaling using 5,7-DHT. This treatment strongly reduced CNO-mediated anti-nociceptive effects in TBI but not uninjured rats. To define the serotonergic receptor(s) required for the CNO-stimulated effects in TBI rats, we administered 5-HT7 (SB-269970) and 5-HT1A (WAY-100635) receptor antagonists but observed no effects. The selective 5-HT2A receptor antagonist ketanserin, however, blocked CNO's effects in the DREADD expressing TBI but not DREADD expressing sham TBI animals. Blockade of alpha-1 adrenergic receptors with prazosin also had no effect after TBI. Descending pain control originating in the PAG is mediated through opioid receptors in uninjured rats. TBI, however, fundamentally alters the descending nociceptive control circuitry such that serotonergic influences predominate, and those are mediated by the 5-HT2A receptor. These results provide further evidence that the PAG is a key target for anti-nociception after TBI.
Collapse
Affiliation(s)
- Karen-Amanda Irvine
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California, USA
- Anesthesiology Service Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Xiao-You Shi
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California, USA
- Anesthesiology Service Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Adam R Ferguson
- Brain and Spinal Injury Center, Department of Neurosurgery, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
- San Francisco Veterans Affairs Healthcare System, San Francisco, California, USA
| | - J David Clark
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, California, USA
- Anesthesiology Service Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| |
Collapse
|
5
|
Park EJ, Truong VL, Jeong WS, Min WK. Brain-Derived Neurotrophic Factor (BDNF) Enhances Osteogenesis and May Improve Bone Microarchitecture in an Ovariectomized Rat Model. Cells 2024; 13:518. [PMID: 38534361 PMCID: PMC10969057 DOI: 10.3390/cells13060518] [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: 01/08/2024] [Revised: 03/09/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) has gained attention as a therapeutic agent due to its potential biological activities, including osteogenesis. However, the molecular mechanisms involved in the osteogenic activity of BDNF have not been fully understood. This study aimed to investigate the action of BDNF on the osteoblast differentiation in bone marrow stromal cells, and its influence on signaling pathways. In addition, to evaluate the clinical efficacy, an in vivo animal study was performed. METHODS Preosteoblast cells (MC3T3-E1), bone marrow-derived stromal cells (ST2), and a direct 2D co-culture system were treated with BDNF. The effect of BDNF on cell proliferation was determined using the CCK-8 assay. Osteoblast differentiation was assessed based on alkaline phosphatase (ALP) activity and staining and the protein expression of multiple osteoblast markers. Calcium accumulation was examined by Alizarin red S staining. For the animal study, we used ovariectomized Sprague-Dawley rats and divided them into BDNF and normal saline injection groups. MicroCT, hematoxylin and eosin (H&E), and tartrate-resistant acid phosphatase (TRAP) stain were performed for analysis. RESULTS BDNF significantly increased ALP activity, calcium deposition, and the expression of osteoblast differentiation-related proteins, such as ALP, osteopontin, etc., in both ST-2 and the MC3T3-E1 and ST-2 co-culture systems. Moreover, the effect of BDNF on osteogenic differentiation was diminished by blocking tropomyosin receptor kinase B, as well as inhibiting c-Jun N-terminal kinase and p38 MAPK signals. Although the animal study results including bone density and histology showed increased osteoblastic and decreased osteoclastic activity, only a portion of parameters reached statistical significance. CONCLUSIONS Our study results showed that BDNF affects osteoblast differentiation through TrkB receptor, and JNK and p38 MAPK signal pathways. Although not statistically significant, the trend of such effects was observed in the animal experiment.
Collapse
Affiliation(s)
- Eugene J. Park
- Department of Orthopedic Surgery, Kyungpook National University Hospital, College of Medicine, Kyungpook National University, Daegu 41566, Republic of Korea;
| | - Van-Long Truong
- Food and Bio-Industry Research Institute, School of Food Science & Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea;
| | - Woo-Sik Jeong
- Food and Bio-Industry Research Institute, School of Food Science & Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea;
| | - Woo-Kie Min
- Department of Orthopedic Surgery, Kyungpook National University Hospital, College of Medicine, Kyungpook National University, Daegu 41566, Republic of Korea;
| |
Collapse
|
6
|
Bharadwaj VN, Sahbaie P, Shi X, Irvine KA, Yeomans DC, Clark JD. Effect of Voluntary Exercise on Endogenous Pain Control Systems and Post-traumatic Headache in Mice. THE JOURNAL OF PAIN 2023; 24:1859-1874. [PMID: 37271350 PMCID: PMC11806938 DOI: 10.1016/j.jpain.2023.05.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/16/2023] [Accepted: 05/30/2023] [Indexed: 06/06/2023]
Abstract
Traumatic brain injury (TBI) can cause acute and chronic pain along with motor, cognitive, and emotional problems. Although the mechanisms are poorly understood, previous studies suggest disruptions in endogenous pain modulation may be involved. Voluntary exercise after a TBI has been shown to reduce some consequences of injury including cognitive impairment. We hypothesized, therefore, that voluntary exercise could augment endogenous pain control systems in a rodent model of TBI. For these studies, we used a closed-head impact procedure in male mice modeling mild TBI. We investigated the effect of voluntary exercise on TBI-induced hindpaw nociceptive sensitization, diffuse noxious inhibitory control failure, and periorbital sensitization after bright light stress, a model of post-traumatic headache. Furthermore, we investigated the effects of exercise on memory, circulating markers of brain injury, neuroinflammation, and spinal cord gene expression. We observed that exercise significantly reduced TBI-induced hindpaw allodynia and periorbital allodynia in the first week following TBI. We also showed that exercise improved the deficits associated with diffuse noxious inhibitory control and reduced bright light stress-induced allodynia up to 2 months after TBI. In addition, exercise preserved memory and reduced TBI-induced increases in spinal BDNF, CXCL1, CXCL2, and prodynorphin expression, all genes previously linked to TBI-induced nociceptive sensitization. Taken together, our observations suggest that voluntary exercise may reduce pain after TBI by reducing TBI-induced changes in nociceptive signaling and preserving endogenous pain control systems. PERSPECTIVE: This article evaluates the effects of exercise on pain-related behaviors in a preclinical model of traumatic brain injury (TBI). The findings show that exercise reduces nociceptive sensitization, loss of diffuse noxious inhibitory control, memory deficits, and spinal nociception-related gene expression after TBI. Exercise may reduce or prevent pain after TBI.
Collapse
Affiliation(s)
- Vimala N Bharadwaj
- Department of Anesthesia, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, California.
| | - Peyman Sahbaie
- Department of Anesthesia, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, California; Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Xiaoyou Shi
- Department of Anesthesia, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, California; Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - Karen-Amanda Irvine
- Department of Anesthesia, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, California; Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| | - David C Yeomans
- Department of Anesthesia, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, California
| | - J David Clark
- Department of Anesthesia, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, California; Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, California
| |
Collapse
|
7
|
Wong KR, Wright DK, Sgro M, Salberg S, Bain J, Li C, Sun M, McDonald SJ, Mychasiuk R, Brady RD, Shultz SR. Persistent Changes in Mechanical Nociception in Rats With Traumatic Brain Injury Involving Polytrauma. THE JOURNAL OF PAIN 2023; 24:1383-1395. [PMID: 36958460 DOI: 10.1016/j.jpain.2023.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 03/12/2023] [Accepted: 03/15/2023] [Indexed: 03/25/2023]
Abstract
Traumatic brain injury (TBI) survivors often experience debilitating consequences. Due to the high impact nature of TBI, patients often experience concomitant peripheral injuries (ie, polytrauma). A common, yet often overlooked, comorbidity of TBI is chronic pain. Therefore, this study investigated how common concomitant peripheral injuries (ie, femoral fracture and muscle crush) can affect long-term behavioral and structural TBI outcomes with a particular focus on nociception. Rats were randomly assigned to 1 of 4 groups: polytrauma (POLY; ie, fracture + muscle crush + TBI), peripheral injury (PERI; ie, fracture + muscle crush + sham TBI), TBI (ie, sham fracture + sham muscle crush + TBI), and sham-injured (SHAM; ie, sham fracture + sham muscle crush + sham TBI). Rats underwent behavioral testing at 3-, 6-, and 11-weeks postinjury, and were then euthanized for postmortem magnetic resonance imaging (MRI). POLY rats had a persisting increase in pain sensitivity compared to all groups on the von Frey test. MRI revealed that POLY rats also had abnormalities in the cortical and subcortical brain structures involved in nociceptive processing. These findings have important implications and provide a foundation for future studies to determine the underlying mechanisms and potential treatment strategies for chronic pain in TBI survivors. PERSPECTIVE: Rats with TBI and concomitant peripheral trauma displayed chronic nociceptive pain and MRI images also revealed damaged brain structures/pathways that are involved in chronic pain development. This study highlights the importance of polytrauma and the affected brain regions for developing chronic pain.
Collapse
Affiliation(s)
- Ker Rui Wong
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - David K Wright
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Marissa Sgro
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Sabrina Salberg
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Jesse Bain
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Crystal Li
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Mujun Sun
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, VIC, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Rhys D Brady
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia; Department of Medicine, The University of Melbourne, Parkville, VIC, Australia; Department of Nursing, Health and Human Services, Vancouver Island University, Nanaimo, BC, Canada.
| |
Collapse
|
8
|
Jeon S, Baik J, Kim J, Lee J, Do W, Kim E, Lee HJ, Kim H. Intrathecal dexmedetomidine attenuates mechanical allodynia through the downregulation of brain-derived neurotrophic factor in a mild traumatic brain injury rat model. Korean J Anesthesiol 2023; 76:56-66. [PMID: 35760392 PMCID: PMC9902181 DOI: 10.4097/kja.22209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/20/2022] [Accepted: 06/23/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND This study evaluated the effects of dexmedetomidine and propofol on brain-derived neurotrophic factor level in the cerebrospinal fluid (c-BDNF) and mechanical allodynia in a mild traumatic brain injury (TBI) rat model. METHODS After fixing the rat's skull on a stereotactic frame under general anesthesia, craniotomy was performed. After impact, 10 µl of drug was injected into the cisterna magna (group S: sham, group D: dexmedetomidine 5 μg/kg, group P: propofol 500 μg/kg, and group T: untreated TBI). The 50% mechanical withdrawal threshold (50% MWT) and c-BDNF level were measured on postoperative days (PODs) 1, 7, and 14. RESULTS The 50% MWT measured on PODs 1, 7, and 14 was lower and the c-BDNF level on POD 1 was higher in group T than in group S. In group D, the c-BDNF level on POD 1 was lower than that in group T and was comparable with that in group S during the whole study period. The 50% MWT of group D was higher than that of group T throughout the postoperative period. In group P, there were no significant differences in the 50% MWT during the entire postoperative period compared with group T; the c-BDNF level was higher than that in group T on POD 1. CONCLUSIONS Intrathecal administration of dexmedetomidine may attenuate TBI-induced mechanical allodynia for up to two weeks post-injury through immediate suppression of c-BDNF in mild TBI rats. The inhibition of c-BDNF expression in the acute phase reduced the occurrence of TBI-induced chronic neuropathic pain.
Collapse
Affiliation(s)
- Soeun Jeon
- Department of Anesthesiology and Pain Medicine, School of Dentistry, Institute for Translational Research in Dentistry, Kyungpook National University, Daegu, Korea
| | - Jiseok Baik
- Department of Anesthesia and Pain Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
- Pusan National University School of Medicine, Busan, Korea
| | - Jisu Kim
- Department of Anesthesia and Pain Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Jiyoon Lee
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Wangseok Do
- Department of Anesthesia and Pain Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Eunsoo Kim
- Department of Anesthesia and Pain Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
- Pusan National University School of Medicine, Busan, Korea
| | - Hyeon Jeong Lee
- Department of Anesthesia and Pain Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
- Pusan National University School of Medicine, Busan, Korea
| | - Haekyu Kim
- Department of Anesthesia and Pain Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
- Pusan National University School of Medicine, Busan, Korea
| |
Collapse
|
9
|
Best KM, Mojena MM, Barr GA, Schmidt HD, Cohen AS. Endogenous Opioid Dynorphin Is a Potential Link between Traumatic Brain Injury, Chronic Pain, and Substance Use Disorder. J Neurotrauma 2022; 39:1-19. [PMID: 34751584 PMCID: PMC8978570 DOI: 10.1089/neu.2021.0063] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Traumatic brain injury (TBI) is a serious public health problem associated with numerous physical and neuropsychiatric comorbidities. Chronic pain is prevalent and interferes with post-injury functioning and quality of life, whereas substance use disorder (SUD) is the third most common neuropsychiatric diagnosis after TBI. Neither of these conditions has a clear mechanistic explanation based on the known pathophysiology of TBI. Dynorphin is an endogenous opioid neuropeptide that is significantly dysregulated after TBI. Both dynorphin and its primary receptor, the ĸ-opioid receptor (KOR), are implicated in the neuropathology of chronic pain and SUD. Here, we review the known roles of dynorphin and KORs in chronic pain and SUDs. We synthesize this information with our current understanding of TBI and highlight potential mechanistic parallels between and across conditions that suggest a role for dynorphin in long-term sequelae after TBI. In pain studies, dynorphin/KOR activation has either antinociceptive or pro-nociceptive effects, and there are similarities between the signaling pathways influenced by dynorphin and those underlying development of chronic pain. Moreover, the dynorphin/KOR system is considered a key regulator of the negative affective state that characterizes drug withdrawal and protracted abstinence in SUD, and molecular and neurochemical changes observed during the development of SUD are mirrored by the pathophysiology of TBI. We conclude by proposing hypotheses and directions for future research aimed at elucidating the potential role of dynorphin/KOR in chronic pain and/or SUD after TBI.
Collapse
Affiliation(s)
- Kaitlin M. Best
- Department of Nursing and Clinical Care Services, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marissa M. Mojena
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Gordon A. Barr
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Psychology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Heath D. Schmidt
- Department of Biobehavioral Health Sciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Akiva S. Cohen
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Address correspondence to: Akiva S. Cohen, PhD, Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, 3615 Civic Center Boulevard, Room 816-I, Philadelphia, PA 19104, USA
| |
Collapse
|
10
|
Do W, Baik J, Jeon S, You CM, Kang D, Jung YH, Lee J, Kim HK. Increased Brain-Derived Neurotrophic Factor Levels in Cerebrospinal Fluid During the Acute Phase in TBI-Induced Mechanical Allodynia in the Rat Model. J Pain Res 2022; 15:229-239. [PMID: 35125890 PMCID: PMC8809523 DOI: 10.2147/jpr.s344110] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/21/2022] [Indexed: 11/23/2022] Open
Abstract
Background The present study aimed to develop a rat model for mechanical allodynia after traumatic brain injury (TBI) and to investigate the expression of brain-derived neurotrophic factor (BDNF) in the cerebrospinal fluid (CSF) using this model. Methods A total of 180 rats were randomly allocated into three groups: a control group (group C), a sham-operated group (group S), and a controlled cortical impact induced TBI group (group T), 60 in each group. Von Frey test was performed to evaluate mechanical withdrawal thresholds. An enzyme-linked immunosorbent assay was performed to quantify BDNF level in CSF. Results The 50% withdrawal thresholds of group T were lower than those of group C and group S at all measuring points except for the preoperative period (P = 0.026, <0.001, and <0.001 for POD1, POD7, and POD14, respectively). The BDNF level of group T was higher than those of group C and group S at POD1 (P = 0.005). Conclusion Upregulation of the BDNF expression in CSF was observed in rats who developed mechanical allodynia on the day after TBI. Based on our findings, to elucidate the relationship between TBI-induced neuropathic pain and BDNF expression in CSF, further research should be carried out through a multifaceted approach to a broad spectrum of pain behavior models.
Collapse
Affiliation(s)
- Wangseok Do
- Department of Anesthesia and Pain Medicine and Biomedical Research Institute Pusan National University Hospital, Busan, Republic of Korea
| | - Jiseok Baik
- Department of Anesthesia and Pain Medicine and Biomedical Research Institute Pusan National University Hospital, Busan, Republic of Korea
- Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Busan, Republic of Korea
- Correspondence: Jiseok Baik, Department of Anesthesia and Pain Medicine and Biomedical Research Institute Pusan National University Hospital, 179 Gudeok-Ro, Seo-gu, Busan, 49241, Republic of Korea, Tel +82-51-240-7499, Fax +82-51-242-7466, Email
| | - Soeun Jeon
- Department of Anesthesia and Pain Medicine and Biomedical Research Institute Pusan National University Hospital, Busan, Republic of Korea
| | - Chang-Min You
- Department of Anesthesia and Pain Medicine and Biomedical Research Institute Pusan National University Hospital, Busan, Republic of Korea
| | - Dahyun Kang
- Department of Anesthesia and Pain Medicine and Biomedical Research Institute Pusan National University Hospital, Busan, Republic of Korea
| | - Young-Hoon Jung
- Department of Anesthesia and Pain Medicine and Biomedical Research Institute Pusan National University Hospital, Busan, Republic of Korea
| | - Jiyoon Lee
- Department of Anesthesia and Pain Medicine, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Hae-Kyu Kim
- Department of Anesthesia and Pain Medicine and Biomedical Research Institute Pusan National University Hospital, Busan, Republic of Korea
- Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
11
|
Naugle KM, Carey C, Evans E, Saxe J, Overman R, White FA. The role of deficient pain modulatory systems in the development of persistent post-traumatic headaches following mild traumatic brain injury: an exploratory longitudinal study. J Headache Pain 2020; 21:138. [PMID: 33272206 PMCID: PMC7712573 DOI: 10.1186/s10194-020-01207-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Post-traumatic headache (PTH) is one of the most common and long-lasting symptoms following mild traumatic brain injury (TBI). However, the pathological mechanisms underlying the development of persistent PTH remain poorly understood. The primary purpose of this prospective pilot study was to evaluate whether early pain modulatory profiles (sensitization and endogenous pain inhibitory capacity) and psychological factors after mild TBI predict the development of persistent PTH in mild TBI patients. METHODS Adult mild TBI patients recruited from Level I Emergency Department Trauma Centers completed study sessions at 1-2 weeks, 1-month, and 4-months post mild TBI. Participants completed the following outcome measures during each session: conditioned pain modulation to measure endogenous pain inhibitory capacity, temporal summation of pain and pressure pain thresholds of the head to measure sensitization of the head, Pain Catastrophizing Scale, Center for Epidemiological Studies - Depression Scale, and a standardized headache survey. Participants were classified into persistent PTH (PPTH) and no-PPTH groups based on the 4-month data. RESULTS The results revealed that mild TBI patients developing persistent PTH exhibited significantly diminished pain inhibitory capacity, and greater depression and pain catastrophizing following injury compared to those who do not develop persistent PTH. Furthermore, logistic regression indicated that headache pain intensity at 1-2 weeks and pain inhibitory capacity on the conditioned pain modulation test at 1-2 weeks predicted persistent PTH classification at 4 months post injury. CONCLUSIONS Overall, the results suggested that persistent PTH is characterized by dysfunctional alterations in endogenous pain modulatory function and psychological processes in the early stages following mild TBI, which likely exacerbate risk for the maintenance of PTH.
Collapse
Affiliation(s)
- Kelly M Naugle
- Department of Kinesiology, School of Health and Human Sciences, Indiana University Purdue University Indianapolis (IUPUI), 901 West New York St., Indianapolis, IN, 46202, USA.
| | - Christopher Carey
- Department of Kinesiology, School of Health and Human Sciences, Indiana University Purdue University Indianapolis (IUPUI), 901 West New York St., Indianapolis, IN, 46202, USA
| | - Eric Evans
- Department of Health Sciences, School of Health and Human Sciences, IUPUI, 250 University Boulevard, Indianapolis, IN, 46202, USA
| | - Jonathan Saxe
- Trauma Department, Ascension St. Vincent Indianapolis Hospital, 2001 W 86th St, Indianapolis, IN, 46260, USA
| | - Ryan Overman
- Department of Neurology, Indiana University School of Medicine, GH 4700 Neur, IN, 46202, Indianapolis, USA
| | - Fletcher A White
- Department of Anesthesia, School of Medicine, Indiana University, 320 West 15th Street, Indianapolis, IN, 46202, USA.,Stark Neuroscience Research Institute, School of Medicine, Indiana University, 320 West 15th Street, Indianapolis, IN, 46202, USA
| |
Collapse
|
12
|
Shultz SR, McDonald SJ, Corrigan F, Semple BD, Salberg S, Zamani A, Jones NC, Mychasiuk R. Clinical Relevance of Behavior Testing in Animal Models of Traumatic Brain Injury. J Neurotrauma 2020; 37:2381-2400. [DOI: 10.1089/neu.2018.6149] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Sandy R. Shultz
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Stuart J. McDonald
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Melbourne, Victoria, Australia
| | - Frances Corrigan
- Department of Anatomy, University of South Australia, Adelaide, South Australia, Australia
| | - Bridgette D. Semple
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Sabrina Salberg
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Akram Zamani
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Nigel C. Jones
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
13
|
Irvine KA, Sahbaie P, Ferguson AR, Clark JD. Loss of diffuse noxious inhibitory control after traumatic brain injury in rats: A chronic issue. Exp Neurol 2020; 333:113428. [PMID: 32745472 PMCID: PMC11793995 DOI: 10.1016/j.expneurol.2020.113428] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/23/2020] [Accepted: 07/25/2020] [Indexed: 02/07/2023]
Abstract
Chronic pain is one of the most challenging and debilitating symptoms to manage after traumatic brain injury (TBI), yet the underlying mechanisms remain elusive. The disruption of normal endogenous pain control mechanisms has been linked to several forms of chronic pain and may play a role in pain after TBI. We hypothesized therefore that dysfunctional descending noradrenergic and serotonergic pain control circuits may contribute to the loss of diffuse noxious inhibitory control (DNIC), a critical endogenous pain control mechanism, weeks to months after TBI. For these studies, the rat lateral fluid percussion model of mild TBI was used along with a DNIC paradigm involving a capsaicin-conditioning stimulus. We observed sustained failure of the DNIC response up to 180-days post injury. We confirmed, that descending α2 adrenoceptor-mediated noradrenergic signaling was critical for endogenous pain inhibition in uninjured rats. However, augmenting descending noradrenergic signaling using reboxetine, a selective noradrenaline reuptake inhibitor, failed to restore DNIC after TBI. Furthermore, blocking serotonin-mediated descending signaling using selective spinal serotonergic fiber depletion with 5, 7-dihydroxytryptamine was also unsuccessful at restoring endogenous pain modulation after TBI. Unexpectedly, increasing descending serotonergic signaling using the selective serotonin reuptake inhibitor escitalopram and the serotonin-norepinephrine reuptake inhibitor duloxetine restored the DNIC response in TBI rats at both 49- and 180- days post injury. Consistent with these observations, spinal serotonergic fiber depletion with 5, 7-dihydroxytryptamine eliminated the effects of escitalopram. Intact α2 adrenoceptor signaling, however, was not required for the serotonin-mediated restoration of DNIC after TBI. These results suggest that TBI causes maladaptation of descending nociceptive signaling mechanisms and changes in the function of both adrenergic and serotonergic circuits. Such changes could predispose those with TBI to chronic pain.
Collapse
Affiliation(s)
- Karen-Amanda Irvine
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, CA 94305, USA; Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, 3801 Miranda Ave (E4-220), Palo Alto, CA 94304, USA.
| | - Peyman Sahbaie
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, CA 94305, USA; Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, 3801 Miranda Ave (E4-220), Palo Alto, CA 94304, USA
| | - Adam R Ferguson
- University of California San Francisco, Brain and Spinal Injury Center, Department Neurosurgery, 1001 Potrero Ave, San Francisco, CA 94110, USA
| | - J David Clark
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, CA 94305, USA; Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, 3801 Miranda Ave (E4-220), Palo Alto, CA 94304, USA
| |
Collapse
|
14
|
Blaze J, Choi I, Wang Z, Umali M, Mendelev N, Tschiffely AE, Ahlers ST, Elder GA, Ge Y, Haghighi F. Blast-Related Mild TBI Alters Anxiety-Like Behavior and Transcriptional Signatures in the Rat Amygdala. Front Behav Neurosci 2020; 14:160. [PMID: 33192359 PMCID: PMC7604767 DOI: 10.3389/fnbeh.2020.00160] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/11/2020] [Indexed: 12/21/2022] Open
Abstract
The short and long-term neurological and psychological consequences of traumatic brain injury (TBI), and especially mild TBI (mTBI) are of immense interest to the Veteran community. mTBI is a common and detrimental result of combat exposure and results in various deleterious outcomes, including mood and anxiety disorders, cognitive deficits, and post-traumatic stress disorder (PTSD). In the current study, we aimed to further define the behavioral and molecular effects of blast-related mTBI using a well-established (3 × 75 kPa, one per day on three consecutive days) repeated blast overpressure (rBOP) model in rats. We exposed adult male rats to the rBOP procedure and conducted behavioral tests for anxiety and fear conditioning at 1-1.5 months (sub-acute) or 12-13 months (chronic) following blast exposure. We also used next-generation sequencing to measure transcriptome-wide gene expression in the amygdala of sham and blast-exposed animals at the sub-acute and chronic time points. Results showed that blast-exposed animals exhibited an anxiety-like phenotype at the sub-acute timepoint but this phenotype was diminished by the chronic time point. Conversely, gene expression analysis at both sub-acute and chronic timepoints demonstrated a large treatment by timepoint interaction such that the most differentially expressed genes were present in the blast-exposed animals at the chronic time point, which also corresponded to a Bdnf-centric gene network. Overall, the current study identified changes in the amygdalar transcriptome and anxiety-related phenotypic outcomes dependent on both blast exposure and aging, which may play a role in the long-term pathological consequences of mTBI.
Collapse
Affiliation(s)
- Jennifer Blaze
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Inbae Choi
- Research and Development Service, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
| | - Zhaoyu Wang
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Michelle Umali
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Natalia Mendelev
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Anna E Tschiffely
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical Research Center, Silver Spring, MD, United States
| | - Stephen T Ahlers
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical Research Center, Silver Spring, MD, United States
| | - Gregory A Elder
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Neurology Service, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States
| | - Yongchao Ge
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Fatemeh Haghighi
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Research and Development Service, James J. Peters Veterans Affairs Medical Center, Bronx, NY, United States.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
15
|
Benemei S, Labastida-Ramírez A, Abramova E, Brunelli N, Caronna E, Diana P, Gapeshin R, Hofacker MD, Maestrini I, Pías EM, Mikulenka P, Tikhonova O, Martelletti P, MaassenVanDenBrink A. Persistent post-traumatic headache: a migrainous loop or not? The preclinical evidence. J Headache Pain 2020; 21:90. [PMID: 32664898 PMCID: PMC7362418 DOI: 10.1186/s10194-020-01135-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 05/25/2020] [Indexed: 12/11/2022] Open
Abstract
Background According to the International Classification of Headache Disorders 3, post-traumatic headache (PTH) attributed to traumatic brain injury (TBI) is a secondary headache reported to have developed within 7 days from head injury, regaining consciousness following the head injury, or discontinuation of medication(s) impairing the ability to sense or report headache following the head injury. It is one of the most common secondary headache disorders, and it is defined as persistent when it lasts more than 3 months. Main body Currently, due to the high prevalence of this disorder, several preclinical studies have been conducted using different animal models of mild TBI to reproduce conditions that engender PTH. Despite representing a simplification of a complex disorder and displaying different limitations concerning the human condition, animal models are still a mainstay to study in vivo the mechanisms of PTH and have provided valuable insight into the pathophysiology and possible treatment strategies. Different models reproduce different types of trauma and have been ideated in order to ensure maximal proximity to the human condition and optimal experimental reproducibility. Conclusion At present, despite its high prevalence, PTH is not entirely understood, and the differential contribution of pathophysiological mechanisms, also observed in other conditions like migraine, has to be clarified. Although facing limitations, animal models are needed to improve understanding of PTH. The knowledge of currently available models is necessary to all researchers who want to investigate PTH and contribute to unravel its mechanisms.
Collapse
Affiliation(s)
- Silvia Benemei
- Health Sciences Department, University of Florence and Headache Centre, Careggi University Hospital, Florence, Italy
| | - Alejandro Labastida-Ramírez
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Dr Molewaterplein 50, 3015 GE, Rotterdam, The Netherlands
| | - Ekaterina Abramova
- Pain Clinic Unit, Department of Anesthesiology, Pirogov City Clinical Hospital, Moscow, Russia
| | | | - Edoardo Caronna
- Neurology Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Paola Diana
- Child Neuropsychiatry Unit, Department of PROMISE, University of Palermo, Palermo, Italy
| | - Roman Gapeshin
- Department of Neurology and Manual Medicine, Pavlov First Saint-Petersburg State Medical University, Saint-Petersburg, Russia
| | - Maxi Dana Hofacker
- Department of Neurology, Headache Centre, Charité Universitatsmedizin Berlin, Berlin, Germany
| | - Ilaria Maestrini
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Enrique Martínez Pías
- Neurology Department, Hospital Clínico Universitario of Valladolid, Valladolid, Spain
| | - Petr Mikulenka
- Department of Neurology, Third Faculty of Medicine, Charles University and University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Olga Tikhonova
- Department of neurology, Kazaryan Clinic of Epileptology and Neurology, Moscow, Russia
| | - Paolo Martelletti
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Antoinette MaassenVanDenBrink
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Dr Molewaterplein 50, 3015 GE, Rotterdam, The Netherlands.
| | | |
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW Posttraumatic headache (PTH) attributed to mild traumatic brain injury is common and debilitating. In up to one-half of those with acute PTH, the PTH becomes persistent (PTH), enduring for longer than 3 months. The high incidence and persistence of PTH necessitate research into PTH pathophysiology and treatment. In this review, recent developments regarding the diagnostic criteria for PTH, the pathophysiology of PTH, and PTH treatment are discussed. RECENT FINDINGS International Classification of Headache Disorders 3 diagnostic criteria for PTH attributed to head trauma require that 'a headache of any type' starts within 7 days of a head injury. PTH is considered 'persistent' when it endures for more than 3 months. Preclinical and human PTH research suggest multiple pathophysiologic mechanisms including genetic influences, neuroinflammation, increased release and inadequate clearance of neuropeptides and neurotransmitters, mast cell degranulation, and brain structural and functional remodeling. Even when it has a phenotype similar to a primary headache, data suggest that PTH is distinct from primary headaches. There is a lack of high-quality evidence for the acute or preventive treatment of PTH. However, results from published studies of conventional headache therapies and newer therapies, such as calcitonin gene-related peptide mAbs and transcranial magnetic stimulation, justify the current and future randomized controlled trials. SUMMARY Evidence points towards a complex pathophysiology for PTH that is at least partially distinct from the primary headaches. Although properly conducted clinical trials of PTH treatment are needed, existing work has provided important data that help to plan these clinical trials. Current and future investigations will help to identify PTH mechanisms, predictors for PTH persistence, therapeutic targets, and evidence-based treatment options.
Collapse
|
17
|
Sahbaie P, Irvine KA, Liang DY, Shi X, Clark JD. Mild Traumatic Brain Injury Causes Nociceptive Sensitization through Spinal Chemokine Upregulation. Sci Rep 2019; 9:19500. [PMID: 31863005 PMCID: PMC6925232 DOI: 10.1038/s41598-019-55739-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 12/02/2019] [Indexed: 11/28/2022] Open
Abstract
High rates of acute and chronic pain are associated with traumatic brain injury (TBI), but mechanisms responsible for the association remain elusive. Recent data suggest dysregulated descending pain modulation circuitry could be involved. Based on these and other observations, we hypothesized that serotonin (5-HT)-dependent activation of spinal CXC Motif Chemokine Receptor 2 (CXCR2) may support TBI-related nociceptive sensitization in a mouse model of mild TBI (mTBI). We observed that systemic 5-HT depletion with p-chlorophenylalanine attenuated mechanical hypersensitivity seen after mTBI. Likewise, selective spinal 5-HT fiber depletion with 5,7-dihydroxytryptamine (5,7-DHT) reduced hypersensitivity after mTBI. Consistent with a role for spinal 5-HT3 serotonin receptors, intrathecal ondansetron administration after TBI dose-dependently attenuated nociceptive sensitization. Also, selective CXCR2 antagonist SCH527123 treatment attenuated mechanical hypersensitivity after mTBI. Furthermore, spinal CXCL1 and CXCL2 mRNA and protein levels were increased after mTBI as were GFAP and IBA-1 markers. Spinal 5,7-DHT application reduced both chemokine expression and glial activation. Our results suggest dual pathways for nociceptive sensitization after mTBI, direct 5-HT effect through 5-HT3 receptors and indirectly through upregulation of chemokine signaling. Designing novel clinical interventions against either the 5-HT3 mediated component or chemokine pathway may be beneficial in treating pain frequently seen in patients after mTBI.
Collapse
Affiliation(s)
- Peyman Sahbaie
- Department of Anesthesia, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, CA, 94305, USA. .,Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, 3801 Miranda Ave (112-A), Palo Alto, CA, 94304, USA.
| | - Karen-Amanda Irvine
- Department of Anesthesia, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, CA, 94305, USA.,Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, 3801 Miranda Ave (112-A), Palo Alto, CA, 94304, USA
| | - De-Yong Liang
- Department of Anesthesia, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, CA, 94305, USA.,Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, 3801 Miranda Ave (112-A), Palo Alto, CA, 94304, USA
| | - Xiaoyou Shi
- Department of Anesthesia, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, CA, 94305, USA.,Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, 3801 Miranda Ave (112-A), Palo Alto, CA, 94304, USA
| | - J David Clark
- Department of Anesthesia, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, CA, 94305, USA.,Anesthesiology Service, Veterans Affairs Palo Alto Health Care System, 3801 Miranda Ave (112-A), Palo Alto, CA, 94304, USA
| |
Collapse
|
18
|
Enhanced descending pain facilitation in acute traumatic brain injury. Exp Neurol 2019; 320:112976. [PMID: 31185197 DOI: 10.1016/j.expneurol.2019.112976] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/25/2019] [Accepted: 06/06/2019] [Indexed: 01/23/2023]
Abstract
Acute and persistent pain are recognized consequences of TBI that can enhance suffering and significantly impair rehabilitative efforts. Both experimental models and clinical studies suggest that TBI may result in an imbalance between descending pain facilitatory and inhibitory pathways. The aim of this study was to assess the role of enhanced descending serotonin-mediated pain facilitation in a rat TBI model using selective spinal serotonergic fiber depletion with 5, 7-dihydroxytryptamine (DHT). We observed significant hindpaw allodynia in TBI rats that was reduced after DHT but not vehicle treatment. Immunohistochemical studies demonstrated profound spinal serotonin depletion in DHT-treated rats. Furthermore, lumbar intrathecal administration of the 5-HT3 receptor antagonist ondansetron at 7 days post-injury (DPI), when hindpaw allodynia was maximal, also attenuated nociceptive sensitization. Additional immunohistochemical analyses of the lumbar spinal cord at 7 DPI revealed a robust bilateral microglial response in the superficial dorsal horns that was significantly reduced with DHT treatment. Furthermore, serotonin depletion also prevented the TBI-induced bilateral increase in c-Fos positive cells within the Rexed laminae I and II of the dorsal horns. These results indicate that in the weeks following TBI, pain may be responsive to 5-HT3 receptor antagonists or other measures which rebalance descending pain modulation.
Collapse
|
19
|
Das M, Mayilsamy K, Tang X, Han JY, Foran E, Willing AE, Mohapatra SS, Mohapatra S. Pioglitazone treatment prior to transplantation improves the efficacy of human mesenchymal stem cells after traumatic brain injury in rats. Sci Rep 2019; 9:13646. [PMID: 31541141 PMCID: PMC6754424 DOI: 10.1038/s41598-019-49428-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 08/19/2019] [Indexed: 12/13/2022] Open
Abstract
Traumatic brain injury is a leading cause of death and disability around the world. So far, drugs are not available to repair brain damage. Human mesenchymal stem cell (hMSC) transplantation therapy is a promising approach, although the inflammatory microenvironment of the injured brain affects the efficacy of transplanted hMSCs. We hypothesize that reducing the inflammation in the cerebral microenvironment by reducing pro-inflammatory chemokines prior to hMSC administration will improve the efficacy of hMSC therapy. In a rat model of lateral fluid percussion injury, combined pioglitazone (PG) and hMSC (combination) treatment showed less anxiety-like behavior and improved sensorimotor responses to a noxious cold stimulus. Significant reduction in brain lesion volume, neurodegeneration, microgliosis and astrogliosis were observed after combination treatment. TBI induced expression of inflammatory chemokine CCL20 and IL1-β were significantly decreased in the combination treatment group. Combination treatment significantly increased brain-derived neurotrophic factor (BDNF) level and subventricular zone (SVZ) neurogenesis. Taken together, reducing proinflammatory cytokine expression in the cerebral tissues after TBI by PG administration and prior to hMSC therapy improves the outcome of the therapy in which BDNF could have a role.
Collapse
Affiliation(s)
- Mahasweta Das
- James A. Haley Veterans Hospital, University of South Florida College of Medicine, Tampa, FL, 33612, USA
- Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL, 33612, USA
| | - Karthick Mayilsamy
- James A. Haley Veterans Hospital, University of South Florida College of Medicine, Tampa, FL, 33612, USA
- Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL, 33612, USA
| | - Xiaolan Tang
- James A. Haley Veterans Hospital, University of South Florida College of Medicine, Tampa, FL, 33612, USA
- Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL, 33612, USA
| | - Jung Yeon Han
- James A. Haley Veterans Hospital, University of South Florida College of Medicine, Tampa, FL, 33612, USA
- Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL, 33612, USA
| | - Elspeth Foran
- James A. Haley Veterans Hospital, University of South Florida College of Medicine, Tampa, FL, 33612, USA
- Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL, 33612, USA
| | - Alison E Willing
- Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, 33612, USA
| | - Shyam S Mohapatra
- James A. Haley Veterans Hospital, University of South Florida College of Medicine, Tampa, FL, 33612, USA
- Department of Internal Medicine, University of South Florida College of Medicine, Tampa, FL, 33612, USA
| | - Subhra Mohapatra
- James A. Haley Veterans Hospital, University of South Florida College of Medicine, Tampa, FL, 33612, USA.
- Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL, 33612, USA.
| |
Collapse
|
20
|
Ashina H, Porreca F, Anderson T, Amin FM, Ashina M, Schytz HW, Dodick DW. Post-traumatic headache: epidemiology and pathophysiological insights. Nat Rev Neurol 2019; 15:607-617. [DOI: 10.1038/s41582-019-0243-8] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2019] [Indexed: 01/01/2023]
|
21
|
Bouferguène S, Lapierre A, Houzé B, Rainville P, Arbour C. Chronic Central Pain Among Community-Dwelling Survivors of Moderate-to-Severe Traumatic Brain Injury: A Quantitative Sensory Testing Study. Biol Res Nurs 2019; 21:519-531. [DOI: 10.1177/1099800419859078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Central pain associated with changes in sensory thresholds is one of the most enduring consequences of major trauma. Yet it remains sparsely studied among community-dwelling survivors of moderate-to-severe traumatic brain injury (TBI). Purpose: To describe and compare thermal and mechanical sensory thresholds in home-based patients with and without central pain after moderate-to-severe TBI with a cohort of healthy controls. Design: Cross-sectional. Method: Thresholds for cold/heat detection, thermal pain, touch, and distorted sensation were gathered using quantitative sensory testing (QST). QST was performed on the painful and contralateral pain-free body regions in TBI participants with pain (TBI-P) and on both forearms in TBI participants without pain (TBI-NP) and healthy controls (HC). Central pain was characterized using the Brief Pain Inventory–Short Form. Results: We tested 16 TBI-P patients, 17 TBI-NP patients, and 16 HC. Mean time since injury for TBI patients was 24 ± 15 months. TBI-P and TBI-NP patients showed significant loss in innocuous mechanical sensitivity compared to HC ( F = 18.929; Bonferroni-adjusted p ≤ .001). Right–left differences in cold pain sensations were significantly larger in TBI-P than in TBI-NP and HC participants ( F = 14.352; Bonferroni-adjusted p ≤ .001). Elevated heat sensitivity thresholds were also observed in TBI-P participants but remained within normal range. Conclusion: Damage to cutaneous mechanoreceptors is a necessary, but not sufficient, condition for the development of chronic central pain following TBI. Damage or incomplete recovery of cutaneous thermoreceptors may be a contributing factor to chronic pain after TBI.
Collapse
Affiliation(s)
- Sabrina Bouferguène
- Research Center and Trauma Division, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada
| | - Alexandra Lapierre
- Research Center and Trauma Division, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada
| | - Bérengère Houzé
- Institut universitaire de gériatrie de Montréal, Montréal, Québec, Canada
| | - Pierre Rainville
- Institut universitaire de gériatrie de Montréal, Montréal, Québec, Canada
| | - Caroline Arbour
- Research Center and Trauma Division, Hôpital du Sacré-Coeur de Montréal, Montréal, Québec, Canada
| |
Collapse
|
22
|
Caro XJ, Winter EF. Letter to the Editor: Reduced intraepidermal nerve fiber density after a sustained increase in insular glutamate: a proof-of-concept study examining the pathogenesis of small fiber pathology in fibromyalgia. Pain Rep 2019; 4:e733. [PMID: 31583349 PMCID: PMC6749923 DOI: 10.1097/pr9.0000000000000733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 01/17/2019] [Accepted: 01/21/2019] [Indexed: 11/16/2022] Open
Affiliation(s)
- Xavier J. Caro
- Southern California Fibromyalgia Research & Treatment Center, Northridge Hospital Medical Center, Northridge, CA, USA
| | - Earl F. Winter
- Southern California Fibromyalgia Research & Treatment Center, Northridge Hospital Medical Center, Northridge, CA, USA
| |
Collapse
|
23
|
Belardo C, Iannotta M, Boccella S, Rubino RC, Ricciardi F, Infantino R, Pieretti G, Stella L, Paino S, Marabese I, Maisto R, Luongo L, Maione S, Guida F. Oral Cannabidiol Prevents Allodynia and Neurological Dysfunctions in a Mouse Model of Mild Traumatic Brain Injury. Front Pharmacol 2019; 10:352. [PMID: 31040777 PMCID: PMC6476923 DOI: 10.3389/fphar.2019.00352] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/21/2019] [Indexed: 01/28/2023] Open
Abstract
Neurological dysfunctions are the most impactful and persistent consequences of traumatic brain injury (TBI). Indeed, previous reports suggest that an association between TBI and chronic pain syndromes, as well anxio-depressive behaviors, tends to be more common in patients with mild forms of TBI. At present, no effective treatment options are available for these symptoms. In the present study, we used a weight drop mild TBI mouse model to investigate the effect of a commercially available 10% Cannabidiol (CBD) oil on both the sensorial and neuropsychiatric dysfunctions associated with mild TBI through behavioral and biomolecular approaches. TBI mice developed chronic pain associated with anxious and aggressive behavior, followed by a late depressive-like behavior and impaired social interaction. Such behaviors were related with specific changes in neurotransmitters release at cortical levels. CBD oral treatment restored the behavioral alterations and partially normalized the cortical biochemical changes. In conclusion, our data show some of the brain modifications probably responsible for the behavioral phenotype associated with TBI and suggest the CBD as a pharmacological tool to improve neurological dysfunctions caused by the trauma.
Collapse
Affiliation(s)
- Carmela Belardo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Monica Iannotta
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Serena Boccella
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Flavia Ricciardi
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Rosmara Infantino
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Gorizio Pieretti
- Department of Plastic Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Salvatore Paino
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Ida Marabese
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Rosa Maisto
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Livio Luongo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Francesca Guida
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
24
|
Belardo C, Iannotta M, Boccella S, Rubino RC, Ricciardi F, Infantino R, Pieretti G, Stella L, Paino S, Marabese I, Maisto R, Luongo L, Maione S, Guida F. Oral Cannabidiol Prevents Allodynia and Neurological Dysfunctions in a Mouse Model of Mild Traumatic Brain Injury. Front Pharmacol 2019. [PMID: 31040777 DOI: 10.3389/fphar.2019.00352/full] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023] Open
Abstract
Neurological dysfunctions are the most impactful and persistent consequences of traumatic brain injury (TBI). Indeed, previous reports suggest that an association between TBI and chronic pain syndromes, as well anxio-depressive behaviors, tends to be more common in patients with mild forms of TBI. At present, no effective treatment options are available for these symptoms. In the present study, we used a weight drop mild TBI mouse model to investigate the effect of a commercially available 10% Cannabidiol (CBD) oil on both the sensorial and neuropsychiatric dysfunctions associated with mild TBI through behavioral and biomolecular approaches. TBI mice developed chronic pain associated with anxious and aggressive behavior, followed by a late depressive-like behavior and impaired social interaction. Such behaviors were related with specific changes in neurotransmitters release at cortical levels. CBD oral treatment restored the behavioral alterations and partially normalized the cortical biochemical changes. In conclusion, our data show some of the brain modifications probably responsible for the behavioral phenotype associated with TBI and suggest the CBD as a pharmacological tool to improve neurological dysfunctions caused by the trauma.
Collapse
Affiliation(s)
- Carmela Belardo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Monica Iannotta
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Serena Boccella
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Flavia Ricciardi
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Rosmara Infantino
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Gorizio Pieretti
- Department of Plastic Surgery, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Salvatore Paino
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Ida Marabese
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Rosa Maisto
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Livio Luongo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Francesca Guida
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
25
|
Abstract
Traumatic brain injury (TBI) is the cause for long-term disability in more than 3 million patients in the US alone, with chronic pain being the most frequently reported complain. To date, predisposing mechanisms for chronic pain in TBI patients are largely unknown. Psychological disorders, including post-traumatic stress disorder, depression and anxiety following TBI are commonly reported comorbidities to post-traumatic pain. Long term consequences can be debilitating and affect quality of life even when the injury is mild. In this review, we present the most commonly reported chronic pain conditions across the spectrum of severity of TBI, mainly focusing on mild TBI. We discuss chronic post- traumatic headaches, widespread pain as well as post-traumatic central pain. We discuss pain in the context of injury severity and military versus civilian populations. We are only starting to understand the biological mechanisms behind post-traumatic pain and associated psychological distress following TBI, with genetic, biochemical and imaging studies pointing to the dopaminergic, neurotrophic factors and the role of Apolipoprotein. Physiological and neurological mechanisms are proposed to partially explain this interaction between post-traumatic pain and psychological distress. Nevertheless, the evidence for the role of structural brain damage remains incomplete and to a large extent debatable, as it is still difficult to establish clear causality between brain trauma and chronic pain. Finally, general aspects of management of chronic pain post-TBI are addressed.
Collapse
|
26
|
Sahbaie P, Tajerian M, Yang P, Irvine KA, Huang TT, Luo J, Wyss-Coray T, Clark JD. Nociceptive and Cognitive Changes in a Murine Model of Polytrauma. THE JOURNAL OF PAIN 2018; 19:1392-1405. [DOI: 10.1016/j.jpain.2018.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 06/12/2018] [Accepted: 06/19/2018] [Indexed: 12/27/2022]
|
27
|
Elliott MB, Ward SJ, Abood ME, Tuma RF, Jallo JI. Understanding the endocannabinoid system as a modulator of the trigeminal pain response to concussion. Concussion 2018; 2:CNC49. [PMID: 30202590 PMCID: PMC6122691 DOI: 10.2217/cnc-2017-0010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/29/2017] [Indexed: 12/29/2022] Open
Abstract
Post-traumatic headache is the most common symptom of postconcussion syndrome and becomes a chronic neurological disorder in a substantial proportion of patients. This review provides a brief overview of the epidemiology of postconcussion headache, research models used to study this disorder, as well as the proposed mechanisms. An objective of this review is to enhance the understanding of how the endogenous cannabinoid system is essential for maintaining the balance of the CNS and regulating inflammation after injury, and in turn making the endocannabinoid system a potential modulator of the trigeminal response to concussion. The review describes the role of endocannabinoid modulation of pain and the potential for use of phytocannabinoids to treat pain, migraine and concussion.
Collapse
Affiliation(s)
- Melanie B Elliott
- Department of Neurosurgery, Vickie & Jack Farber Institute for Neuroscience Thomas Jefferson University, PA 19107, USA.,Department of Neurosurgery, Vickie & Jack Farber Institute for Neuroscience Thomas Jefferson University, PA 19107, USA
| | - Sara J Ward
- Department of Pharmacology, Lewis Katz School of Medicine, Temple University, PA 19140, USA.,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, PA 19140, USA
| | - Mary E Abood
- Department of Anatomy & Cell Biology, Lewis Katz School of Medicine, Temple University, PA 19140, USA.,Department of Anatomy & Cell Biology, Lewis Katz School of Medicine, Temple University, PA 19140, USA
| | - Ronald F Tuma
- Department of Physiology Lewis Katz School of Medicine, Temple University, PA 19140, USA.,Department of Physiology Lewis Katz School of Medicine, Temple University, PA 19140, USA
| | - Jack I Jallo
- Department of Neurosurgery, Vickie & Jack Farber Institute for Neuroscience Thomas Jefferson University, PA 19107, USA.,Department of Neurosurgery, Vickie & Jack Farber Institute for Neuroscience Thomas Jefferson University, PA 19107, USA
| |
Collapse
|
28
|
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]
|
29
|
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
|
30
|
Ko IG, Kim SE, Hwang L, Jin JJ, Kim CJ, Kim BK, Kim H. Late starting treadmill exercise improves spatial leaning ability through suppressing CREP/BDNF/TrkB signaling pathway following traumatic brain injury in rats. J Exerc Rehabil 2018; 14:327-334. [PMID: 30018914 PMCID: PMC6028205 DOI: 10.12965/jer.1836248.124] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 05/15/2018] [Indexed: 12/28/2022] Open
Abstract
Traumatic brain injury (TBI) causes deficit in spatial learning and memory function. Physical activity ameliorates neurological dysfunction after TBI. We investigated the effect of late starting treadmill exercise on spatial learning ability in relation with cAMP-response element binding protein (CREB)/brain-derived neurotrophic factor (BDNF) signaling pathway using TBI rats. For this study, radial 8-arm maze test, TUNEL (terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling) staining, caspase-3 immunohistochemistry, and western blot for Bax, Bcl-2, BDNF, tyrosine kinase B (TrkB), CREB, and phosphorylated CREP (p-CREB) were performed. TBI was induced by an electromagnetic-controlled cortical impact. The rats in the exercise groups were scheduled to run on a treadmill for 30 min once a day for 8 weeks starting 3 weeks after TBI. TBI impaired spatial learning ability and increased caspase-3 expression in the hippocampal dentate gyrus. TBI enhanced Bax expression and suppressed Bcl-2 expression in the hip-pocampus. TBI increased BDNF and TrkB expressions, resulted in the enhancement of p-CREB/CREB ratio in the hippocampus. However, treadmill exercise improved spatial learning ability, decreased caspase-3 expression, suppressed Bax expression, and increased Bcl-2 expression. Treadmill exercise alleviated TBI-induced over-expression of BDNF and TrkB, which suppressed phosphorylation of CREB in the hippocampus. In the present study, late starting treadmill exercise improved spatial learning ability through suppressing TBI-induced activation of CREB/BDNF/TrkB signaling pathway after TBI.
Collapse
Affiliation(s)
- Il-Gyu Ko
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea.,Kohwang Medical Research Institute, Kyung Hee University, Seoul, Korea
| | - Sung-Eun Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea.,Kohwang Medical Research Institute, Kyung Hee University, Seoul, Korea
| | - Lakkyong Hwang
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea.,Kohwang Medical Research Institute, Kyung Hee University, Seoul, Korea
| | - Jun-Jang Jin
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea.,Kohwang Medical Research Institute, Kyung Hee University, Seoul, Korea
| | - Chang-Ju Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea.,Kohwang Medical Research Institute, Kyung Hee University, Seoul, Korea
| | - Bo-Kyun Kim
- Department of Emergency Technology, College of Health Science, Gachon University, Incheon, Korea
| | - Hong Kim
- Department of Oriental Sports Medicine, College of Biomedical Science, Daegu Haany University, Gyeongsan, Korea
| |
Collapse
|
31
|
Nervous system delivery of antilysophosphatidic acid antibody by nasal application attenuates mechanical allodynia after traumatic brain injury in rats. Pain 2018; 158:2181-2188. [PMID: 29028747 DOI: 10.1097/j.pain.0000000000001019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Lysophosphatidic acid (LPA) is a bioactive lipid that impacts neurological outcomes after neurotrauma by inhibiting neuroregeneration, promoting inflammation, and contributing to behavioral deficits. Blocking LPA signaling with a novel anti-LPA monoclonal antibody (mAb) is neuroprotective after traumatic brain injury (TBI) if given to injured animals whose blood-brain barrier (BBB) has been compromised. It is hypothesized that the anti-LPA mAb could improve chronic pain initiated by TBI. However, poor brain penetration after systemic application of the antibody makes access to the central nervous system (CNS) problematic in situations where the BBB is intact. Our experiments investigated whether intranasal delivery of the anti-LPA mAb could bypass the BBB, allowing for direct entry of the antibody to certain areas of the CNS. When the humanized anti-LPA mAb, LT3114, was intranasally applied to injured rats within 30 minutes after mild TBI using the central lateral percussion model, enzyme-linked immunospecific assay and immunohistochemistry demonstrated antibody uptake to several areas in the CNS, including the area of cortical injury, the corpus callosum, cerebellum, and the subventricular region. Compared with control rats that received LT3114 but no TBI, TBI rats demonstrated significantly higher concentrations of intranasally administered LT3114 antibody in some tissues. In behavioral studies, a significant attenuation of mechanical allodynia after TBI was observed in the anti-LPA treatment group (P = 0.0079), when compared with vehicle controls within 14 days after TBI. These results suggest that intranasal application of the anti-LPA antibody directly accesses CNS sites involved in TBI-related pain and that this access attenuates pain sequelae to the neurotrauma.
Collapse
|
32
|
Irvine KA, Sahbaie P, Liang DY, Clark JD. Traumatic Brain Injury Disrupts Pain Signaling in the Brainstem and Spinal Cord. J Neurotrauma 2018; 35:1495-1509. [PMID: 29373948 DOI: 10.1089/neu.2017.5411] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Chronic pain is a common consequence of traumatic brain injury (TBI) that can increase the suffering of a patient and pose a significant challenge to rehabilitative efforts. Unfortunately, the mechanisms linking TBI to pain are poorly understood, and specific treatments for TBI-related pain are still lacking. Our laboratory has shown that TBI causes pain sensitization in areas distant to the site of primary injury, and that changes in spinal gene expression may underlie this sensitization. The aim of this study was to examine the roles that pain modulatory pathways descending from the brainstem play in pain after TBI. Deficiencies in one type of descending inhibition, diffuse noxious inhibitory control (DNIC), have been suggested to be responsible for the development of chronic pain by allowing excess and uncontrolled afferent nociceptive inputs. Here we expand our knowledge of pain after TBI in two ways: (1) by outlining the neuropathology in pain-related centers of the brain and spinal cord involved in DNIC using the rat lateral fluid percussion (LFP) model of TBI, and (2) by evaluating the effects of a potent histone acetyl transferase inhibitor, anacardic acid (AA), on LFP-induced pain behaviors and neuropathology when administered for several days after TBI. The results revealed that TBI induces transient mechanical allodynia and a chronic persistent loss of DNIC. Further, while short-term AA treatment can block acute nociceptive sensitization and some early neuropathological changes, this treatment neither prevented the loss of DNIC nor did it alter long-term neuropathological changes in the brain or spinal cord.
Collapse
Affiliation(s)
- Karen-Amanda Irvine
- 1 Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System , Palo Alto, California.,2 Department of Anesthesia, Perioperative Medicine and Pain, Stanford University , Stanford, California
| | - Peyman Sahbaie
- 1 Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System , Palo Alto, California.,2 Department of Anesthesia, Perioperative Medicine and Pain, Stanford University , Stanford, California
| | - De-Yong Liang
- 1 Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System , Palo Alto, California.,2 Department of Anesthesia, Perioperative Medicine and Pain, Stanford University , Stanford, California
| | - J David Clark
- 1 Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System , Palo Alto, California.,2 Department of Anesthesia, Perioperative Medicine and Pain, Stanford University , Stanford, California
| |
Collapse
|
33
|
The Neurotoxin DSP-4 Induces Hyperalgesia in Rats that is Accompanied by Spinal Oxidative Stress and Cytokine Production. Neuroscience 2018; 376:13-23. [PMID: 29421433 DOI: 10.1016/j.neuroscience.2018.01.058] [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] [Received: 07/11/2017] [Revised: 01/26/2018] [Accepted: 01/29/2018] [Indexed: 12/30/2022]
Abstract
Central neuropathic pain (CNP) a significant problem for many people, is not well-understood and difficult to manage. Dysfunction of the central noradrenergic system originating in the locus coeruleus (LC) may be a causative factor in the development of CNP. The LC is the major noradrenergic nucleus of the brain and plays a significant role in central modulation of nociceptive neurotransmission. Here, we examined CNS pathophysiological changes induced by intraperitoneal administration of the neurotoxin DSP-4 (N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine hydrochloride). Administration of DSP-4 decreased levels of norepinephrine in spinal tissue and cerebrospinal fluid (CSF) and led to the development of thermal and mechanical hyperalgesia over 21 days, that was reversible with morphine. Hyperalgesia was accompanied by significant increases in noradrenochrome (oxidized norepinephrine) and expression of 4-hydroxynonenal in CSF and spinal cord tissue respectively at day 21, indicative of oxidative stress. In addition, spinal levels of pro-inflammatory cytokines (interleukins 6 and 17A, tumor necrosis factor-α), as well as the anti-inflammatory cytokine interleukin10 were also significantly elevated at day 21, indicating that an inflammatory response occurred. The inflammatory effect of DSP-4 presented in this study that includes oxidative stress may be particularly useful in elucidating mechanisms of CNP in inflammatory disease states.
Collapse
|
34
|
Meidahl AC, Eisenried A, Klukinov M, Cao L, Tzabazis AZ, Yeomans DC. Intranasal Oxytocin Attenuates Reactive and Ongoing, Chronic Pain in a Model of Mild Traumatic Brain Injury. Headache 2017; 58:545-558. [PMID: 29266199 DOI: 10.1111/head.13248] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2017] [Indexed: 11/30/2022]
Abstract
BACKGROUND Approximately 1.7 million Americans sustain a traumatic brain injury (TBI) each year and chronic pain is a common complication. OBJECTIVE We studied the effects of intranasally administered oxytocin as a potential treatment for chronic pain in an animal model of mild TBI. METHODS The lateral fluid percussion model of mild TBI was chosen for this purpose and after exposure to mild TBI the rats (n = 12) developed hind paw and facial allodynia compared to sham animals (n = 6). Oxytocin or a vehicle was afterwards administered intranasally and reactive pain was assessed by hind paw and facial von Frey testing. Some animals received the oxytocin receptor antagonist, atosiban, in addition to oxytocin/vehicle treatment (n = 12). The effect of oxytocin on ongoing and spontaneous pain was examined through conditioned place preference testing. To determine whether the effects of intranasal oxytocin could be attributed to delivery via the peripheral blood stream, some TBI animals received an intravenous injection of the same oxytocin dose that was given intranasally. ELISA immunoassays were carried out (n = 6) to measure concentrations of oxytocin in the trigeminal ganglia, pons, spinal cord, and olfactory bulb after intranasal administration and evaluate the most likely route of entry. RESULTS These studies confirmed that the fluid percussion model can be used to study post-TBI facial allodynia. Oxytocin attenuated both reactive and spontaneous, ongoing non-reactive pain following mild TBI for at least 3-4 hours after intranasal administration by binding to OT or VA1-receptors most likely by a peri-trigeminal nerve mediated uptake. CONCLUSIONS Intranasal oxytocin attenuates measures of reactive and non-reactive pain in a model of mild TBI and may represent a novel treatment for chronic pain in TBI patients.
Collapse
Affiliation(s)
- Anders C Meidahl
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, CA, USA
| | - Andreas Eisenried
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, CA, USA
| | - Michael Klukinov
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, CA, USA
| | - Longhui Cao
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, CA, USA
| | - Alexander Z Tzabazis
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, CA, USA
| | - David C Yeomans
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, School of Medicine, Stanford, CA, USA
| |
Collapse
|
35
|
Irvine KA, Clark JD. Chronic Pain After Traumatic Brain Injury: Pathophysiology and Pain Mechanisms. PAIN MEDICINE 2017; 19:1315-1333. [DOI: 10.1093/pm/pnx153] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Karen-Amanda Irvine
- Veterans Affairs Palo Alto Health Care System, Anesthesiology Service, Palo Alto, California
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - J David Clark
- Veterans Affairs Palo Alto Health Care System, Anesthesiology Service, Palo Alto, California
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
36
|
Meidahl AC, Klukinov M, Tzabazis AZ, Sorensen JC, Yeomans DC. Nasal application of HSV encoding human preproenkephalin blocks craniofacial pain in a rat model of traumatic brain injury. Gene Ther 2017; 24:482-486. [PMID: 28682314 DOI: 10.1038/gt.2017.55] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 04/06/2017] [Accepted: 06/14/2017] [Indexed: 12/18/2022]
Abstract
According to Centers for Disease Control and Prevention, each year, an estimated 1.7 million Americans sustain a traumatic brain injury (TBI), which frequently leads to chronic craniofacial pain. In this study we examine a gene therapy approach to the treatment of post-TBI craniofacial neuropathic pain using nasal application of a herpes simplex virus (HSV)-based vector expressing human proenkephalin (SHPE) to target the trigeminal ganglia. Mild TBI was induced in rats by the use of a modified fluid percussion model. Two days after mild TBI, following the development of facial mechanical allodynia, animals received either an intranasal application of vehicle or recombinant HSV encoding human preproenkephalin or lacZ reporter gene encoding control vector (SHZ.1). Compared with baseline response thresholds, mild TBI in SHZ.1 or vehicle-treated animals induced a robust craniofacial allodynia lasting at least 45 days. On the other hand, nasal SHPE application 2 days post-TBI attenuated facial allodynia, reaching significance by day 4-7 and maintaining this effect throughout the duration of the experiment. Immunohistochemical examination revealed strong expression of human proenkephalin in trigeminal ganglia of SHPE, but not SHZ.1-treated rats. This study demonstrates that intranasal administration of HSV-based gene vectors may be a viable, non-invasive means of treating chronic craniofacial pain, including post-TBI pain.
Collapse
Affiliation(s)
- A C Meidahl
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA.,Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark
| | - M Klukinov
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - A Z Tzabazis
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - J C Sorensen
- Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark
| | - D C Yeomans
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
37
|
Guida F, Boccella S, Iannotta M, De Gregorio D, Giordano C, Belardo C, Romano R, Palazzo E, Scafuro MA, Serra N, de Novellis V, Rossi F, Maione S, Luongo L. Palmitoylethanolamide Reduces Neuropsychiatric Behaviors by Restoring Cortical Electrophysiological Activity in a Mouse Model of Mild Traumatic Brain Injury. Front Pharmacol 2017; 8:95. [PMID: 28321191 PMCID: PMC5337754 DOI: 10.3389/fphar.2017.00095] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 02/14/2017] [Indexed: 11/25/2022] Open
Abstract
Traumatic brain injury (TBI) represents a major public health problem, which is associated with neurological dysfunction. In severe or moderate cases of TBI, in addition to its high mortality rate, subjects may encounter diverse behavioral dysfunctions. Previous reports suggest that an association between TBI and chronic pain syndromes tends to be more common in patients with mild forms of brain injury. Despite causing minimal brain damage, mild TBI (mTBI) often leads to persistent psychologically debilitating symptoms, which can include anxiety, various forms of memory and learning deficits, and depression. At present, no effective treatment options are available for these symptoms, and little is known about the complex cellular activity affecting neuronal activity that occurs in response to TBI during its late phase. Here, we used a mouse model to investigate the effect of Palmitoylethanolamide (PEA) on both the sensorial and neuropsychiatric dysfunctions associated with mTBI through behavioral, electrophysiological, and biomolecular approaches. Fourteen-day mTBI mice developed anxious, aggressive, and reckless behavior, whilst depressive-like behavior and impaired social interactions were observed from the 60th day onward. Altered behavior was associated with changes in interleukin 1 beta (IL-1β) expression levels and neuronal firing activity in the medial prefrontal cortex. Compared with vehicle, PEA restored the behavioral phenotype and partially normalized the biochemical and functional changes occurring at the supraspinal level. In conclusion, our findings reveal some of the supraspinal modifications responsible for the behavioral alterations associated with mTBI and suggest PEA as a pharmacological tool to ameliorate neurological dysfunction induced by the trauma.
Collapse
Affiliation(s)
- Francesca Guida
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN)Naples, Italy; Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle RicerchePozzuoli, Italy
| | - Serena Boccella
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Monica Iannotta
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Danilo De Gregorio
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Catia Giordano
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Carmela Belardo
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Rosaria Romano
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Enza Palazzo
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Maria A Scafuro
- Department of Anesthesiology, Surgery and Emergency, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Nicola Serra
- Department of Radiology, Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Vito de Novellis
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN) Naples, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN)Naples, Italy; Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle RicerchePozzuoli, Italy
| | - Livio Luongo
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Università degli Studi della Campania "Luigi Vanvitelli" (Ex SUN)Naples, Italy; Endocannabinoid Research Group, Institute of Biomolecular Chemistry, Consiglio Nazionale delle RicerchePozzuoli, Italy; Young Against Pain (YAP) Italian Group, NaplesItaly
| |
Collapse
|
38
|
Tyburski AL, Cheng L, Assari S, Darvish K, Elliott MB. Frequent mild head injury promotes trigeminal sensitivity concomitant with microglial proliferation, astrocytosis, and increased neuropeptide levels in the trigeminal pain system. J Headache Pain 2017; 18:16. [PMID: 28176234 PMCID: PMC5296267 DOI: 10.1186/s10194-017-0726-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 01/18/2017] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Frequent mild head injuries or concussion along with the presence of headache may contribute to the persistence of concussion symptoms. METHODS In this study, the acute effects of recovery between mild head injuries and the frequency of injuries on a headache behavior, trigeminal allodynia, was assessed using von Frey testing up to one week after injury, while histopathological changes in the trigeminal pain pathway were evaluated using western blot, ELISA and immunohistochemistry. RESULTS: A decreased recovery time combined with an increased mild closed head injury (CHI) frequency results in reduced trigeminal allodynia thresholds compared to controls. The repetitive CHI group with the highest injury frequency showed the greatest reduction in trigeminal thresholds along with greatest increased levels of calcitonin gene-related peptide (CGRP) in the trigeminal nucleus caudalis. Repetitive CHI resulted in astrogliosis in the central trigeminal system, increased GFAP protein levels in the sensory barrel cortex, and an increased number of microglia cells in the trigeminal nucleus caudalis. CONCLUSIONS Headache behavior in rats is dependent on the injury frequency and recovery interval between mild head injuries. A worsening of headache behavior after repetitive mild head injuries was concomitant with increases in CGRP levels, the presence of astrocytosis, and microglia proliferation in the central trigeminal pathway. Signaling between neurons and proliferating microglia in the trigeminal pain system may contribute to the initiation of acute headache after concussion or other traumatic brain injuries.
Collapse
Affiliation(s)
- Ashley L Tyburski
- Department of Neurosurgery, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA, 19107, USA
| | - Lan Cheng
- Department of Neurosurgery, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA, 19107, USA
| | | | | | - Melanie B Elliott
- Department of Neurosurgery, Thomas Jefferson University, 1020 Locust Street, Philadelphia, PA, 19107, USA.
| |
Collapse
|
39
|
Moye LS, Pradhan AA. From blast to bench: A translational mini-review of posttraumatic headache. J Neurosci Res 2017; 95:1347-1354. [PMID: 28151589 DOI: 10.1002/jnr.24001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 11/07/2016] [Accepted: 11/28/2016] [Indexed: 12/25/2022]
Abstract
Current events within the military and professional sports have resulted in an increased recognition of the long-term and debilitating consequences of traumatic brain injury. Mild traumatic brain injury accounts for the majority of head injuries, and posttraumatic headache is the most common adverse effect. It is estimated that between 30% to 90% of traumatic brain injuries result in posttraumatic headache, and for a significant number of people this headache disorder can continue for up to and over a year post injury. Often, the most severe and chronic posttraumatic headache has a migraine-like phenotype and is difficult to resolve. In this review we discuss the preclinical findings from animal models of posttraumatic headache. We also describe potential mechanisms by which traumatic brain injury leads to chronic posttraumatic headache, including neuroinflammatory mediators and migraine-associated neuropeptides. There are surprisingly few preclinical studies that have investigated overlapping mechanisms between posttraumatic headache and migraine, especially considering the prevalence and debilitating nature of posttraumatic headache. Given this context, posttraumatic headache is a field with many emerging opportunities for growth. The frequency of posttraumatic headache in the general and military population is rising, and further preclinical research is required to understand, ameliorate, and treat this disabling disorder. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Laura S Moye
- Department of Psychiatry, University of Illinois at Chicago
| | | |
Collapse
|
40
|
|
41
|
Liang DY, Shi X, Liu P, Sun Y, Sahbaie P, Li WW, Yeomans DC, Clark JD. The Chemokine Receptor CXCR2 Supports Nociceptive Sensitization after Traumatic Brain Injury. Mol Pain 2017; 13:1744806917730212. [PMID: 28845733 PMCID: PMC5593214 DOI: 10.1177/1744806917730212] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 07/07/2017] [Accepted: 07/15/2017] [Indexed: 11/16/2022] Open
Abstract
Abstract Chronic pain after traumatic brain injury (TBI) is very common, but the mechanisms linking TBI to pain and the pain-related interactions of TBI with peripheral injuries are poorly understood. Chemokine receptors play an important role in both pain and brain injury. In the current work, we pursued the hypothesis that the epigenetically regulated CXC chemokine receptor 2 (CXCR2) is a crucial modulator of nociceptive sensitization induced by TBI. For these studies, we used the rat lateral fluid percussion model of TBI. Histone actyltransferase activity was blocked using anacardic acid beginning immediately following injury, or delayed for seven days prior to administration. The selective CXCR2 antagonist SCH527123 administered systemically or intrathecally was used to probe the role of chemokine signaling on mechanical hindpaw sensitization after TBI. The expression of the CXCR2 receptor was accomplished using real-time PCR, immunohistochemistry, and Western blotting, while epigenetic regulation was assessed using chromatin immunoprecipitation assay. The spinal levels of several pain-related mediators including CXCL1, an endogenous ligand for CXCR2, as well as brain-derived neurotrophic factor and prodynorphin were measured by enzyme-linked immunosorbent assay. We observed that anacardic acid potently blocked and reversed mechanical hindpaw sensitization after TBI. The same drug was able to prevent the upregulation of CXCR2 after TBI, but did not affect the spinal expression of other pain mediators. On the other hand, both systemically and intrathecally administered SCH527123 reversed hindpaw allodynia after TBI. Most of the spinal CXCR2 appeared to be expressed by spinal cord neurons. Chromatin immunoprecipitation experiments demonstrated TBI-enhanced association of the CXCR2 promoter with acetylated-H3K9 histone protein that was also reversible using anacardic acid. Taken together, our findings suggested that TBI causes the upregulation of spinal CXCR2 through an epigenetic mechanism ultimately supporting nociceptive sensitization. The use of CXCR2 antagonists may, therefore, be useful in pain resulting from TBI.
Collapse
Affiliation(s)
- De-Yong Liang
- Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiaoyou Shi
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Peng Liu
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuan Sun
- Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Peyman Sahbaie
- Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Wen-Wu Li
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - David C Yeomans
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - J David Clark
- Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
42
|
Liang DY, Sahbaie P, Sun Y, Irvine KA, Shi X, Meidahl A, Liu P, Guo TZ, Yeomans DC, Clark JD. TBI-induced nociceptive sensitization is regulated by histone acetylation. IBRO Rep 2016; 2:14-23. [PMID: 30135929 PMCID: PMC6084866 DOI: 10.1016/j.ibror.2016.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 12/21/2016] [Indexed: 12/16/2022] Open
Abstract
Chronic pain after traumatic brain injury (TBI) is very common, but the mechanisms linking TBI to pain and the pain-related interactions of TBI with peripheral injuries are poorly understood. In these studies we pursued the hypothesis that TBI pain sensitization is associated with histone acetylation in the rat lateral fluid percussion model. Some animals received hindpaw incisions in addition to TBI to mimic polytrauma. Neuropathological analysis of brain tissue from sham and TBI animals revealed evidence of bleeding, breakdown of the blood brain barrier, in the cortex, hippocampus, thalamus and other structures related to pain signal processing. Mechanical allodynia was measured in these animals for up to eight weeks post-injury. Inhibitors of histone acetyltransferase (HAT) and histone deacetylase (HDAC) were used to probe the role of histone acetylation in such pain processing. We followed serum markers including glial fibrillary acidic protein (GFAP), neuron-specific enolase 2 (NSE) myelin basic protein (MBP) and S100β to gauge TBI injury severity. Our results showed that TBI caused mechanical allodynia in the hindpaws of the rats lasting several weeks. Hindpaws contralateral to TBI showed more rapid and profound sensitization than ipsilateral hindpaws. The inhibition of HAT using curcumin 50 mg/kg s.c reduced mechanical sensitization while the HDAC inhibitor suberoylanilide hydroxamic acid 50 mg/kg i.p. prolonged sensitization in the TBI rats. Immunohistochemical analyses of spinal cord tissue localized changes in the level of acetylation of the H3K9 histone mark to dorsal horn neurons. Taken together, these findings demonstrate that TBI induces sustained nociceptive sensitization, and changes in spinal neuronal histone proteins may play an important role. Pain after traumatic brain injury (TBI) is common and often persistent. Using the rat lateral fluid percussion model, it was observed that hindpaw allodynia is present for three weeks after TBI. Damage to pain processing areas of the brain can be demonstrated after TBI. Agents regulating the epigenetic acetylation of histone proteins modified the time course of TBI-induced allodynia.
Collapse
Affiliation(s)
- De-Yong Liang
- Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, 3801 Miranda Ave, 94304, USA.,Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Peyman Sahbaie
- Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, 3801 Miranda Ave, 94304, USA.,Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Yuan Sun
- Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, 3801 Miranda Ave, 94304, USA.,Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Karen-Amanda Irvine
- Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, 3801 Miranda Ave, 94304, USA.,Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Xiaoyou Shi
- Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, 3801 Miranda Ave, 94304, USA.,Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Anders Meidahl
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Peng Liu
- Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, 3801 Miranda Ave, 94304, USA
| | - Tian-Zhi Guo
- Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, 3801 Miranda Ave, 94304, USA
| | - David C Yeomans
- Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - J David Clark
- Department of Anesthesiology, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, 3801 Miranda Ave, 94304, USA.,Department of Anesthesiology, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
43
|
Bree D, Levy D. Development of CGRP-dependent pain and headache related behaviours in a rat model of concussion: Implications for mechanisms of post-traumatic headache. Cephalalgia 2016; 38:246-258. [PMID: 27899434 DOI: 10.1177/0333102416681571] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background and objective Posttraumatic headache (PTH) is one of the most common, debilitating and difficult symptoms to manage after a mild traumatic brain injury, or concussion. However, the mechanisms underlying PTH remain elusive, in part due to the lack of a clinically relevant animal model. Here, we characterized for the first time, headache and pain-related behaviours in a rat model of concussion evoked by a mild closed head injury (mCHI) - the major type of military and civilian related trauma associated with PTH - and tested responses to current and novel headache therapies. Methods Concussion was induced in adult male rats using a weight-drop device. Characterization of headache and pain related behaviours included assessment of cutaneous tactile pain sensitivity, using von Frey monofilaments, and ongoing pain using the conditioned place preference or aversion (CPP/CPA) paradigms. Sensitivity to headache/migraine triggers was tested by exposing rats to low-dose glyceryl trinitrate (GTN). Treatments included acute systemic administration of sumatriptan and chronic systemic administration of a mouse anti-CGRP monoclonal antibody. Results Concussed rats developed cephalic tactile pain hypersensitivity that was resolved by two weeks post-injury and was ameliorated by treatment with sumatriptan or anti-CGRP monoclonal antibody. Sumatriptan also produced CPP seven days post mCHI, but not in sham animals. Following the resolution of the concussion-evoked cephalic hypersensitivity, administration of GTN produced a renewed and pronounced cephalic pain hypersensitivity that was inhibited by sumatriptan or anti-CGRP antibody treatment as well as a CGRP-dependent CPA. GTN had no effect in sham animals. Conclusions Concussion leads to the development of headache and pain-related behaviours, in particular sustained enhanced responses to GTN, that are mediated through a CGRP-dependent mechanism. Treatment with anti-CGRP antibodies may be a useful approach to treat PTH.
Collapse
Affiliation(s)
- Dara Bree
- 1 Departments of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.,2 Harvard Medical School, Boston, MA, USA
| | - Dan Levy
- 1 Departments of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.,2 Harvard Medical School, Boston, MA, USA
| |
Collapse
|
44
|
Allitt BJ, Iva P, Yan EB, Rajan R. Hypo-excitation across all cortical laminae defines intermediate stages of cortical neuronal dysfunction in diffuse traumatic brain injury. Neuroscience 2016; 334:290-308. [PMID: 27530700 DOI: 10.1016/j.neuroscience.2016.08.018] [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: 05/13/2016] [Revised: 07/06/2016] [Accepted: 08/08/2016] [Indexed: 01/18/2023]
Abstract
Traumatic brain injury (TBI) is a major cause of morbidity and mortality world-wide and can result in persistent cognitive, sensory and behavioral dysfunction. Understanding the time course of TBI-induced pathology is essential to effective treatment outcomes. We induced TBI in rats using an impact acceleration method and tested for sensorimotor skill and sensory sensitivity behaviors for two weeks to find persistently poor outcomes post-injury. At two weeks post-injury we made high resolution extracellular recordings from barrel cortex neurons, to simple and complex whisker deflections. We found that the supragranular suppression of neural firing (compared to normal) previously seen in the immediate post-TBI aftermath had spread to include suppression of input and infragranular layers at two weeks post-injury; thus, there was suppression of whisker-driven firing rates in all cortical layers to both stimulus types. Further, there were abnormalities in temporal response patterns such that in layers 3-5 there was a temporal broadening of response patterns in response to both whisker deflection stimulus types and in L2 a narrowing of temporal patterns in response to the complex stimulus. Thus, at two weeks post-TBI, supragranular hypo-excitation has evolved to include deep cortical layers likely as a function of progressive atrophy and neurodegeneration. These results are consistent with the hypothesis that TBI alters the delicate excitatory/inhibitory balance in cortex and likely contributes to temporal broadening of responses and restricts the ability to code for complex sensory stimuli.
Collapse
Affiliation(s)
- Benjamin J Allitt
- Department of Physiology, Monash University, Clayton, VIC, Australia.
| | - Pippa Iva
- Department of Physiology, Monash University, Clayton, VIC, Australia.
| | - Edwin B Yan
- Department of Physiology, Monash University, Clayton, VIC, Australia.
| | - Ramesh Rajan
- Department of Physiology, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
45
|
Rowe RK, Ellis GI, Harrison JL, Bachstetter AD, Corder GF, Van Eldik LJ, Taylor BK, Marti F, Lifshitz J. Diffuse traumatic brain injury induces prolonged immune dysregulation and potentiates hyperalgesia following a peripheral immune challenge. Mol Pain 2016; 12:12/0/1744806916647055. [PMID: 27178244 PMCID: PMC4955995 DOI: 10.1177/1744806916647055] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/30/2016] [Indexed: 01/09/2023] Open
Abstract
Background Nociceptive and neuropathic pain occurs as part of the disease process after traumatic brain injury (TBI) in humans. Central and peripheral inflammation, a major secondary injury process initiated by the traumatic brain injury event, has been implicated in the potentiation of peripheral nociceptive pain. We hypothesized that the inflammatory response to diffuse traumatic brain injury potentiates persistent pain through prolonged immune dysregulation. Results To test this, adult, male C57BL/6 mice were subjected to midline fluid percussion brain injury or to sham procedure. One cohort of mice was analyzed for inflammation-related cytokine levels in cortical biopsies and serum along an acute time course. In a second cohort, peripheral inflammation was induced seven days after surgery/injury with an intraplantar injection of carrageenan. This was followed by measurement of mechanical hyperalgesia, glial fibrillary acidic protein and Iba1 immunohistochemical analysis of neuroinflammation in the brain, and flow cytometric analysis of T-cell differentiation in mucosal lymph. Traumatic brain injury increased interleukin-6 and chemokine ligand 1 levels in the cortex and serum that peaked within 1–9 h and then resolved. Intraplantar carrageenan produced mechanical hyperalgesia that was potentiated by traumatic brain injury. Further, mucosal T cells from brain-injured mice showed a distinct deficiency in the ability to differentiate into inflammation-suppressing regulatory T cells (Tregs). Conclusions We conclude that traumatic brain injury increased the inflammatory pain associated with cutaneous inflammation by contributing to systemic immune dysregulation. Regulatory T cells are immune suppressors and failure of T cells to differentiate into regulatory T cells leads to unregulated cytokine production which may contribute to the potentiation of peripheral pain through the excitation of peripheral sensory neurons. In addition, regulatory T cells are identified as a potential target for therapeutic rebalancing of peripheral immune homeostasis to improve functional outcome and decrease the incidence of peripheral inflammatory pain following traumatic brain injury.
Collapse
Affiliation(s)
- Rachel K Rowe
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA Phoenix Veteran Affairs Healthcare System, Phoenix, AZ, USA
| | - Gavin I Ellis
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Jordan L Harrison
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA Arizona State University, Tempe, AZ, USA
| | - Adam D Bachstetter
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Gregory F Corder
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Linda J Van Eldik
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, USA Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Bradley K Taylor
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Francesc Marti
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Jonathan Lifshitz
- BARROW Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA Phoenix Veteran Affairs Healthcare System, Phoenix, AZ, USA Arizona State University, Tempe, AZ, USA
| |
Collapse
|
46
|
Daiutolo BV, Tyburski A, Clark SW, Elliott MB. Trigeminal Pain Molecules, Allodynia, and Photosensitivity Are Pharmacologically and Genetically Modulated in a Model of Traumatic Brain Injury. J Neurotrauma 2015; 33:748-60. [PMID: 26472135 DOI: 10.1089/neu.2015.4087] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The pain-signaling molecules, nitric oxide synthase (NOS) and calcitonin gene-related peptide (CGRP), are implicated in the pathophysiology of post-traumatic headache (PTH) as they are for migraine. This study assessed the changes of inducible NOS (iNOS) and its cellular source in the trigeminal pain circuit, as well as the relationship between iNOS and CGRP after controlled cortical impact (CCI) injury in mice. The effects of a CGRP antagonist (MK8825) and sumatriptan on iNOS messenger RNA (mRNA) and protein were compared to vehicle at 2 weeks postinjury. Changes in CGRP levels in the trigeminal nucleus caudalis (TNC) in iNOS knockouts with CCI were compared to wild-type (WT) mice at 3 days and 2 weeks post injury. Trigeminal allodynia and photosensitivity were measured. MK8825 and sumatriptan increased allodynic thresholds in CCI groups compared to vehicle (p < 0.01), whereas iNOS knockouts were not different from WT. Photosensitivity was attenuated in MK8825 mice and iNOS knockouts compared to WT (p < 0.05). MK8825 and sumatriptan reduced levels of iNOS mRNA and iNOS immunoreactivity in the TNC and ganglia (p < 0.01). Differences in iNOS cellular localization were found between the trigeminal ganglia and TNC. Although the knockout of iNOS attenuated CGRP at 3 days (p < 0.05), it did not reduce CGRP at 2 weeks. CGRP immunoreactivity was found in the meningeal layers post-CCI, while negligible in controls. Findings support the importance of interactions between CGRP and iNOS in mediating allodynia, as well as the individual roles in photosensitivity. Mitigating prolonged increases in CGRP may be a promising intervention for treating acute PTH.
Collapse
Affiliation(s)
- Brittany V Daiutolo
- Department of Neurological Surgery, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Ashley Tyburski
- Department of Neurological Surgery, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Shannon W Clark
- Department of Neurological Surgery, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Melanie B Elliott
- Department of Neurological Surgery, Thomas Jefferson University , Philadelphia, Pennsylvania
| |
Collapse
|