1
|
Alrasheed AS, Alqadhibi MA, Khoja RH, Alayyaf AS, Alhumoudi DS, Aldawlan MI, Alghanmi BO, Almutairi FS, Bin-Mahfooz MA, Altalhi LA, Aldanyowi SN, Aleid AM, Alessa AA. Emerging therapies for immunomodulation in traumatic brain injury: A systematic review and meta-analysis. Surg Neurol Int 2024; 15:327. [PMID: 39372991 PMCID: PMC11450791 DOI: 10.25259/sni_502_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 08/12/2024] [Indexed: 10/08/2024] Open
Abstract
Background Traumatic brain injury (TBI) represents a significant global health burden, often leading to significant morbidity and mortality. Mounting evidence underscores the intricate involvement of dysregulated immune responses in TBI pathophysiology, highlighting the potential for immunomodulatory interventions to mitigate secondary injury cascades and enhance patient outcomes. Despite advancements in treatment modalities, optimizing therapeutic strategies remains a critical challenge in TBI management. To address this gap, this systematic review and meta-analysis aimed to rigorously evaluate the efficacy and safety of emerging immunomodulatory therapies in the context of TBI. Methods We searched electronic databases such as PubMed, Scopus, Web of Science and CENTRAL for relevant studies investigating the efficacy of immunomodulatory therapies in TBI that were meticulously selected for inclusion. Two independent reviewers meticulously performed data extraction and quality assessment, adhering to predefined criteria. Both randomized controlled trials (RCTs) and observational studies reporting clinically relevant outcomes, such as mortality rates, the Glasgow coma scale, and adverse events, were meticulously scrutinized. Meta-analysis techniques were employed to assess treatment effects across studies quantitatively and analyzed using the Review Manager software (version 5.2). Results Fourteen studies (n = 1 observational and n = 13 RCTs) were included in our study. Meta-analysis showed no significant overall mortality difference, but erythropoietin (EPO) significantly reduced mortality (odds ratio = 0.49; 95% confidence interval: 0.31-0.78, P = 0.002). The adverse event meta-analysis revealed no significant differences. Conclusion Immunomodulatory therapies did not significantly affect overall mortality, but EPO demonstrated promising results. Adverse events did not significantly differ from controls. Further research is warranted to refine TBI treatment protocols.
Collapse
Affiliation(s)
| | | | - Rammaz Hussam Khoja
- Department of Surgery, College of Medicine, Taibah University, Madinah, Saudi Arabia
| | - Abdulaziz Saad Alayyaf
- Department of Surgery, College of Medicine, Prince Sattam bin Abdulaziz University, Riyadh, Saudi Arabia
| | - Duaa Saleh Alhumoudi
- Department of Surgery, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Mubarak Ibrahim Aldawlan
- Department of Surgery, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | | | | | - Mohammed Ali Bin-Mahfooz
- Department of Surgery, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Lina Abdulrahim Altalhi
- Department of Surgery, College of Medicine and Medical Science, Arabian Gulf University, Manama, Bahrain
| | - Saud Nayef Aldanyowi
- Department of Surgery, College of Medicine, King Faisal University, AlAhsa, Saudi Arabia
| | | | | |
Collapse
|
2
|
Comparan HDM, Khaliq A, Frota LM, Pomar-Forero D, Ahmad B, Marnet E, Teixeira FJP, Thomas A, Patel P, Brunkal H, Singireddy S, Lucke-Wold B, Maciel CB, Busl KM. Cyclooxygenase 2 Inhibitors for Headache After Elective Cranial Neurosurgery: Results from a Systematic Review of Efficacy of Cyclooxygenase 2 Inhibitors for Headache After Acute Brain Injury. Neurocrit Care 2024:10.1007/s12028-024-02114-y. [PMID: 39266865 DOI: 10.1007/s12028-024-02114-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 08/22/2024] [Indexed: 09/14/2024]
Abstract
Headache management after acute brain injury (ABI) is challenging. Although opioids are commonly used, selective cyclooxygenase 2 inhibitors (COXIBs) may be promising alternatives. However, concerns about cardiovascular effects and bleeding risk have limited their use. We aimed at summarizing available data on efficacy of COXIBs for headache management following ABI. A systematic review was conducted through MEDLINE and Embase for articles published through September 2023 (PROSPERO identifier: CRD42022320453). No language filters were applied to the initial searches. Interventional or observational studies and systematic reviews assessing efficacy of COXIBs for headache in adults with ABI were eligible. Article selection was performed by two independent reviewers using DistillerSR. Descriptive statistics were used for data analysis, and meta-analysis was unfeasible because of study heterogeneity. Of 3190 articles identified, 6 studies met inclusion criteria: 4 randomized controlled trials and 2 retrospective cohort studies, all conducted in elective cranial neurosurgical patients (total N = 738) between 2006 and 2022. Five studies used COXIBs in the intervention group only. Of the six studies, four found a reduction in overall pain scores in the intervention group, whereas one showed improvement only at 6 h postoperatively, and one did not find significant differences. Pain scores decreased between 4 and 15%, the largest shift being from moderate to mild severity. Three studies found an overall opioid use reduction throughout hospitalization in the intervention group, whereas one reported a reduction at 12 h postoperatively only. Opioid consumption decreased between 9 and 90%. Two studies found a decrease in hospital length of stay by ~ 1 day in the intervention group. The one study reporting postoperative hemorrhage found a statistically nonsignificant 3% reduction in the intervention group. COXIBs may serve as opioid-sparing adjunctive analgesics for headache control after elective cranial surgery. Limited or no literature exists for other forms of ABI, and additional safety data remain to be elucidated.
Collapse
Affiliation(s)
- Hector David Meza Comparan
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Anum Khaliq
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Luciola Martins Frota
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Daniela Pomar-Forero
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Bakhtawar Ahmad
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Erica Marnet
- Department of Medicine, Bridgeport Hospital, Bridgeport, CT, 06610, USA
| | - Fernanda J P Teixeira
- Department of Neurology, University of Miami and Jackson Memorial Hospital, Miami, FL, 33130, USA
| | - Anita Thomas
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Priyank Patel
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Haley Brunkal
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Saanvi Singireddy
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Carolina B Maciel
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, FL, 32611, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06520, USA
- Department of Neurology, University of Utah, Salt Lake City, UT, 84132, USA
| | - Katharina M Busl
- Division of Neurocritical Care, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, 32611, USA.
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
3
|
Zahoor M, Farhat SM, Khan S, Ahmed T. Daidzin improves neurobehavioral outcome in rat model of traumatic brain injury. Behav Brain Res 2024; 472:115158. [PMID: 39047874 DOI: 10.1016/j.bbr.2024.115158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 07/13/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
Traumatic brain injury (TBI) is associated with the etiology of multiple neurological disorders, including neurodegeneration, leading to various cognitive deficits. Daidzin (obtained from kudzu root and soybean leaves) is known for its neuroprotective effects through multiple mechanisms. This study aimed to investigate the pharmacological effects of Daidzin on sensory, and biochemical parameters, cognitive functions, anxiety, and depressive-like behaviors in the TBI rat model. Rats were divided into four groups (Control, TBI, TBI + Ibuprofen (30 mg/kg), and TBI + Daidzin (5 mg/kg)). Rats were subjected to TBI by dropping a 200 g rod from a height of 26 cm, resulting in an impact force of 0.51 J on the exposed crania. Ibuprofen (30 mg/kg) was used as a positive control reference/standard drug and Daidzin (5 mg/kg) as the test drug. Neurological severity score (NSS) assessment was done to determine the intactness of sensory and motor responses. Brain tissue edema and acetylcholine levels were determined in the cortex and hippocampus. Cognitive functions such as hippocampus-dependent memory, novel object recognition, exploration, depressive and anxiety-like behaviors were measured. Treatment with Daidzin improved NSS, reduced hippocampal and cortical edema, and improved levels of acetylcholine in TBI-induced rats. Furthermore, Daidzin treatment improved hippocampus-dependent memory, exploration behavior, and novel object recognition while reducing depressive and anxiety-like behavior. Our study revealed that Daidzin has a therapeutic potential comparable to Ibuprofen and can offer neuroprotection and enhanced cognitive and behavioral outcomes in rats after TBI.
Collapse
Affiliation(s)
- Maryam Zahoor
- Neurobiology Laboratory, Department of Biomedicine, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Sector H-12, Islamabad 44000, Pakistan
| | - Syeda Mehpara Farhat
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi 46000, Pakistan
| | - Salman Khan
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, Pakistan
| | - Touqeer Ahmed
- Neurobiology Laboratory, Department of Biomedicine, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Sector H-12, Islamabad 44000, Pakistan.
| |
Collapse
|
4
|
Boucher ML, Conley G, Morriss NJ, Ospina-Mora S, Qiu J, Mannix R, Meehan WP. Time-Dependent Long-Term Effect of Memantine following Repetitive Mild Traumatic Brain Injury. J Neurotrauma 2024; 41:e1736-e1758. [PMID: 38666723 DOI: 10.1089/neu.2023.0423] [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: 05/16/2024] Open
Abstract
Repetitive mild traumatic brain injury (rmTBI, e.g., sports concussions) may be associated with both acute and chronic symptoms and neurological changes. Despite the common occurrence of these injuries, therapeutic strategies are limited. One potentially promising approach is N-methyl-D-aspartate receptor (NMDAR) blockade to alleviate the effects of post-injury glutamatergic excitotoxicity. Initial pre-clinical work using the NMDAR antagonist, memantine, suggests that immediate treatment following rmTBI improves a variety of acute outcomes. It remains unclear (1) whether acute memantine treatment has long-term benefits and (2) whether delayed treatment following rmTBI is beneficial, which are both clinically relevant concerns. To test this, animals were subjected to rmTBI via a weight drop model with rotational acceleration (five hits in 5 days) and randomized to memantine treatment immediately, 3 months, or 6 months post-injury, with a treatment duration of one month. Behavioral outcomes were assessed at 1, 4, and 7 months post-injury. Neuropathological outcomes were characterized at 7 months post-injury. We observed chronic changes in behavior (anxiety-like behavior, motor coordination, spatial learning, and memory), as well as neuroinflammation (microglia, astrocytes) and tau phosphorylation (T231). Memantine treatment, either immediately or 6 months post-injury, appears to confer greater rescue of neuroinflammatory changes (microglia) than vehicle or treatment at the 3-month time point. Although memantine is already being prescribed chronically to address persistent symptoms associated with rmTBI, this study represents the first evidence of which we are aware to suggest a small but durable effect of memantine treatment in mild, concussive injuries. This effect suggests that memantine, although potentially beneficial, is insufficient to treat all aspects of rmTBI alone and should be combined with other therapeutic agents in a multi-therapy approach, with attention given to the timing of treatment.
Collapse
Affiliation(s)
- Masen L Boucher
- Division of Emergency Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | | | - Nicholas J Morriss
- University of Rochester School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, New York, USA
| | | | - Jianhua Qiu
- Division of Emergency Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Rebekah Mannix
- Division of Emergency Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - William P Meehan
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Division of Sports Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- The Micheli Center for Sports Injury Prevention, Waltham, Massachusetts, USA
| |
Collapse
|
5
|
Leon-Martinez D, Bank TC, Lundsberg LS, Culhane J, Silasi M, Son M, Partridge C, Reddy UM, Hoffman MK, Merriam AA. Does Antenatal Progesterone Administration Modify the Risk of Neonatal Intraventricular Hemorrhage? Am J Perinatol 2024; 41:e46-e52. [PMID: 35436803 DOI: 10.1055/a-1827-6712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
OBJECTIVE Progesterone administration has been associated with improved neurological outcomes following traumatic brain injury in adults. However, studies examining the effect of progesterone on the risk of neonatal intraventricular hemorrhage (IVH) are inconsistent. We sought to determine if maternal administration of intramuscular 17-α-hydroxyprogesterone caproate (17-OHPC) is associated with decreased rates of IVH in infants born before 32-weeks gestation. STUDY DESIGN This is a retrospective study of liveborn singleton deliveries between 20- and 32-weeks gestation at two large academic medical centers from January 1, 2012 to August 30, 2020. Data were extracted from hospital electronic medical record data warehouses using standardized definitions and billing and diagnosis codes. We evaluated receipt of 17-OHPC in the antepartum period and diagnosis of IVH (grade I-IV, per Volpe classification) during the neonatal delivery hospitalization encounter. Bivariate and multivariate analyses were performed to examine the association between 17-OHPC and neonatal IVH adjusting for potential confounders. Odds ratio (ORs) and 95% confidence intervals (CIs) were presented. RESULTS Among 749 neonates born between 20- and 32-week gestation, 140 (18.7%) of their mothers had received antenatal 17-OHPC and 148 (19.8%) were diagnosed with IVH after birth. No significant association was observed between maternal 17-OHPC and neonatal IVH in unadjusted (OR 1.14, 95% CI 0.72-1.78) or adjusted analyses (adjusted odds ratio 1.14, 95% CI 0.71-1.84). Independent of exposure to 17-OHPC, as expected, infants born <28-weeks gestation or those with very low birthweight (<1,500 g) were at an increased risk of IVH (OR 2.32, 95% CI 1.55-3.48 and OR 2.19, 95% CI 1.09-4.38, respectively). CONCLUSION Antenatal maternal 17-OHPC administration was not associated with the risk of neonatal IVH. Further research may be warranted to determine whether timing, route of delivery, and duration of progesterone therapy impact rates of neonatal IVH. KEY POINTS · This study aimed to compare the frequency of intraventricular hemorrhage in preterm neonates exposed to antenatal 17-α-hydroxyprogesterone caproate to those not exposed.. · In neonates born at <32-weeks gestation, maternal use of progesterone is not associated with the risk of intraventricular hemorrhage.. · In contrast to preclinical and adult data, this study suggests that progesterone exposure is not associated with the prevention of neonatal brain injury..
Collapse
Affiliation(s)
- Daisy Leon-Martinez
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Tracy C Bank
- Department of Obstetrics and Gynecology, Christiana Care Health System, Newark, Delaware
| | - Lisbet S Lundsberg
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Jennifer Culhane
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Michelle Silasi
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Moeun Son
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Caitlin Partridge
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Uma M Reddy
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Matthew K Hoffman
- Department of Obstetrics and Gynecology, Christiana Care Health System, Newark, Delaware
| | - Audrey A Merriam
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
6
|
Hummel R, Dorochow E, Zander S, Ritter K, Hahnefeld L, Gurke R, Tegeder I, Schäfer MKE. Valproic Acid Treatment after Traumatic Brain Injury in Mice Alleviates Neuronal Death and Inflammation in Association with Increased Plasma Lysophosphatidylcholines. Cells 2024; 13:734. [PMID: 38727269 PMCID: PMC11083124 DOI: 10.3390/cells13090734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/15/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024] Open
Abstract
The histone deacetylase inhibitor (HDACi) valproic acid (VPA) has neuroprotective and anti-inflammatory effects in experimental traumatic brain injury (TBI), which have been partially attributed to the epigenetic disinhibition of the transcription repressor RE1-Silencing Transcription Factor/Neuron-Restrictive Silencer Factor (REST/NRSF). Additionally, VPA changes post-traumatic brain injury (TBI) brain metabolism to create a neuroprotective environment. To address the interconnection of neuroprotection, metabolism, inflammation and REST/NRSF after TBI, we subjected C57BL/6N mice to experimental TBI and intraperitoneal VPA administration or vehicle solution at 15 min, 1, 2, and 3 days post-injury (dpi). At 7 dpi, TBI-induced an up-regulation of REST/NRSF gene expression and HDACi function of VPA on histone H3 acetylation were confirmed. Neurological deficits, brain lesion size, blood-brain barrier permeability, or astrogliosis were not affected, and REST/NRSF target genes were only marginally influenced by VPA. However, VPA attenuated structural damage in the hippocampus, microgliosis and expression of the pro-inflammatory marker genes. Analyses of plasma lipidomic and polar metabolomic patterns revealed that VPA treatment increased lysophosphatidylcholines (LPCs), which were inversely associated with interleukin 1 beta (Il1b) and tumor necrosis factor (Tnf) gene expression in the brain. The results show that VPA has mild neuroprotective and anti-inflammatory effects likely originating from favorable systemic metabolic changes resulting in increased plasma LPCs that are known to be actively taken up by the brain and function as carriers for neuroprotective polyunsaturated fatty acids.
Collapse
Affiliation(s)
- Regina Hummel
- Department of Anesthesiology, University Medical Center Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (R.H.); (K.R.)
| | - Erika Dorochow
- Institute of Clinical Pharmacology, Medical Faculty, Goethe-University Frankfurt, 60596 Frankfurt am Main, Germany; (E.D.); (L.H.); (R.G.)
| | - Sonja Zander
- Department of Anesthesiology, University Medical Center Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (R.H.); (K.R.)
| | - Katharina Ritter
- Department of Anesthesiology, University Medical Center Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (R.H.); (K.R.)
| | - Lisa Hahnefeld
- Institute of Clinical Pharmacology, Medical Faculty, Goethe-University Frankfurt, 60596 Frankfurt am Main, Germany; (E.D.); (L.H.); (R.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Fraunhofer Cluster of Excellence for Immune-Mediated Diseases, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Robert Gurke
- Institute of Clinical Pharmacology, Medical Faculty, Goethe-University Frankfurt, 60596 Frankfurt am Main, Germany; (E.D.); (L.H.); (R.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Fraunhofer Cluster of Excellence for Immune-Mediated Diseases, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Medical Faculty, Goethe-University Frankfurt, 60596 Frankfurt am Main, Germany; (E.D.); (L.H.); (R.G.)
| | - Michael K. E. Schäfer
- Department of Anesthesiology, University Medical Center Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (R.H.); (K.R.)
- Focus Program Translational Neurosciences (FTN), Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
- Research Center for Immunotherapy (FZI), Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| |
Collapse
|
7
|
Comparan HDM, Khaliq A, Frota LM, Pomar-Forero D, Ahmad B, Marnet E, Teixeira FJP, Thomas A, Patel P, Brunkal H, Singireddy S, Lucke-Wold B, Maciel CB, Busl KM. Efficacy of Cyclooxygenase-2 Inhibitors for Headache in Acute Brain Injury: A Systematic Review. RESEARCH SQUARE 2024:rs.3.rs-4232407. [PMID: 38659820 PMCID: PMC11042422 DOI: 10.21203/rs.3.rs-4232407/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Background Headache management after acute brain injury (ABI) is challenging. While opioids are commonly used, selective cyclooxygenase-2 inhibitors (COXIBs) may be promising alternatives. However, concerns about cardiovascular effects and bleeding risk have limited their use. We aimed at summarizing available data on efficacy of COXIBs for headache management following ABI. Methods A systematic review was conducted through MEDLINE and Embase for articles published through 09/2023 (PROSPERO CRD42022320453). No language filters were applied to the initial searches. Interventional or observational studies and systematic reviews assessing efficacy of COXIBs for headache in adults with ABI were eligible. Article selection was performed by two independent reviewers using Distiller SR®. Descriptive statistics were used for data analysis, while meta-analysis was unfeasible due to study heterogeneity. Results Of 3190 articles identified, six studies met inclusion criteria: four randomized controlled trials and two retrospective cohort studies, all conducted in neurosurgical patients (total n=738) between 2006-2022. Five studies used COXIBs in the intervention group only. Of the six studies, four found a reduction in overall pain scores in the intervention group, while one showed improvement only at 6 hours postoperative, and one did not find significant differences. Pain scores decreased between 4-15%, the largest shift being from moderate to mild severity. Three studies found an overall opioid use reduction throughout hospitalization in the intervention group, while one reported a reduction at 12 hours postoperative only. Opioid consumption decreased between 9-90%. Two studies found a decrease in hospital-length-of-stay by ~1 day in the intervention group. The one study reporting postoperative hemorrhage found a statistically non-significant 3% reduction in the intervention group. Conclusions In adults with ABI, COXIBs may serve as opioid-sparing adjunctive analgesics for headache control, with limited but pointed data to indicate efficacy in the post-neurosurgical setting. However, further safety data remains to be elucidated.
Collapse
Affiliation(s)
- Hector David Meza Comparan
- Department of Neurology, Division of Neurocritical Care, University of Florida, College of Medicine, Gainesville, FL 32611, USA
| | - Anum Khaliq
- Department of Neurology, Division of Neurocritical Care, University of Florida, College of Medicine, Gainesville, FL 32611, USA
| | - Luciola Martins Frota
- Department of Neurology, Division of Neurocritical Care, University of Florida, College of Medicine, Gainesville, FL 32611, USA
| | - Daniela Pomar-Forero
- Department of Neurology, Division of Neurocritical Care, University of Florida, College of Medicine, Gainesville, FL 32611, USA
| | - Bakhtawar Ahmad
- Department of Neurology, Division of Neurocritical Care, University of Florida, College of Medicine, Gainesville, FL 32611, USA
| | - Erica Marnet
- Department of Medicine, Bridgeport Hospital, Bridgeport, CT, USA 06610
| | - Fernanda J P Teixeira
- Department of Neurology, University of Miami/Jackson Memorial Hospital, FL, USA, 33130
| | - Anita Thomas
- Department of Neurology, Division of Neurocritical Care, University of Florida, College of Medicine, Gainesville, FL 32611, USA
| | - Priyank Patel
- Department of Neurology, Division of Neurocritical Care, University of Florida, College of Medicine, Gainesville, FL 32611, USA
| | - Haley Brunkal
- Department of Neurology, Division of Neurocritical Care, University of Florida, College of Medicine, Gainesville, FL 32611, USA
| | - Saanvi Singireddy
- Department of Neurology, Division of Neurocritical Care, University of Florida, College of Medicine, Gainesville, FL 32611, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, College of Medicine, Gainesville, FL 32611, USA
| | - Carolina B Maciel
- Department of Neurology, Division of Neurocritical Care, University of Florida, College of Medicine, Gainesville, FL 32611, USA; Department of Neurosurgery, University of Florida, College of Medicine, Gainesville, FL 32611, USA; Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Neurology, University of Utah, Salt Lake City, UT 84132, USA
| | - Katharina M Busl
- Department of Neurology, Division of Neurocritical Care, University of Florida, College of Medicine, Gainesville, FL 32611, USA; Department of Neurosurgery, University of Florida, College of Medicine, Gainesville, FL 32611, USA
| |
Collapse
|
8
|
Wang L, Zhang Y, Huang M, Yuan Y, Liu X. RIP3 in Necroptosis: Underlying Contributions to Traumatic Brain Injury. Neurochem Res 2024; 49:245-257. [PMID: 37743445 DOI: 10.1007/s11064-023-04038-z] [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: 07/18/2023] [Revised: 09/07/2023] [Accepted: 09/20/2023] [Indexed: 09/26/2023]
Abstract
Traumatic brain injury (TBI) is a global public safety issue that poses a threat to death, characterized by high fatality rates, severe injuries and low recovery rates. There is growing evidence that necroptosis regulates the pathophysiological processes of a variety of diseases, particularly those affecting the central nervous system. Thus, moderate necroptosis inhibition may be helpful in the management of TBI. Receptor-interacting protein kinase (RIP) 3 is a key mediator in the necroptosis, and its absence helps restore the microenvironment at the injured site and improve cognitive impairment after TBI. In this report, we review different domains of RIP3, multiple analyses of necroptosis, and associations between necroptosis and TBI, RIP3, RIP1, and mixed lineage kinase domain-like. Next, we elucidate the potential involvement of RIP3 in TBI and highlight how RIP3 deficiency enhances neuronal function.
Collapse
Affiliation(s)
- Lvxia Wang
- School of Life and Environmental Sciences, Shaoxing University, Zhejiang, China
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, China
| | - Yong Zhang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, China
| | - Min Huang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, China
| | - Yiling Yuan
- Department of Biosciences, Durham University, Durham, UK
| | - Xuehong Liu
- School of Life and Environmental Sciences, Shaoxing University, Zhejiang, China.
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Zhejiang, China.
| |
Collapse
|
9
|
Celorrio M, Shumilov K, Friess SH. Gut microbial regulation of innate and adaptive immunity after traumatic brain injury. Neural Regen Res 2024; 19:272-276. [PMID: 37488877 PMCID: PMC10503601 DOI: 10.4103/1673-5374.379014] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/27/2023] [Accepted: 05/08/2023] [Indexed: 07/26/2023] Open
Abstract
Acute care management of traumatic brain injury is focused on the prevention and reduction of secondary insults such as hypotension, hypoxia, intracranial hypertension, and detrimental inflammation. However, the imperative to balance multiple clinical concerns simultaneously often results in therapeutic strategies targeted to address one clinical concern causing unintended effects in other remote organ systems. Recently the bidirectional communication between the gastrointestinal tract and the brain has been shown to influence both the central nervous system and gastrointestinal tract homeostasis in health and disease. A critical component of this axis is the microorganisms of the gut known as the gut microbiome. Changes in gut microbial populations in the setting of central nervous system disease, including traumatic brain injury, have been reported in both humans and experimental animal models and can be further disrupted by off-target effects of patient care. In this review article, we will explore the important role gut microbial populations play in regulating brain-resident and peripheral immune cell responses after traumatic brain injury. We will discuss the role of bacterial metabolites in gut microbial regulation of neuroinflammation and their potential as an avenue for therapeutic intervention in the setting of traumatic brain injury.
Collapse
Affiliation(s)
- Marta Celorrio
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Kirill Shumilov
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Stuart H. Friess
- Department of Pediatrics, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|
10
|
Jussen D, Amoruso E, Kempski O, Lucia K, Czabanka M, Ringel F, Alessandri B. Early Onset of Rapid Lesion Growth in an Acute Subdural Hematoma Model in Rats. World Neurosurg 2023; 178:e578-e584. [PMID: 37532019 DOI: 10.1016/j.wneu.2023.07.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023]
Abstract
OBJECTIVE Acute subdural hematoma (ASDH) leads to the highest mortality rates of all head injuries with secondary brain damage playing a pivotal role in terms of morbidity and mortality. In patients with ASDH, a delay in surgery leads to disproportional mortality. The benefit of (very) early therapy is therefore, a target of ongoing research. As the process of delayed brain damage in ASDH has not yet been described, this study therefore aimed to examine secondary lesion growth in an experimental rat model of ASDH to define the ideal timing for testing potential neuroprotective therapies. METHODS Cerebral blood flow was monitored during ASDH induction with 300 μl of autologous blood. Lesion growth was characterized using Hematoxylin-Eosin- , Cresyl-Violet-, and Fluoro-Jade B-staining for early signs of neuronal degeneration. Histological evaluations were performed between 15 minutes and 24 hours after ASDH. RESULTS There was a significant reduction of cerebral blood flow after ASDH. Fluoro-Jade B-positive cells were visible 15 minutes after ASDH in the lesioned hemisphere. Nonlinear growth of lesion volume from 3.7 ± 0.4 mm3 to 17.5 ± 0.6 mm3 was observed at 24 hours in Hematoxylin-Eosin-staining. CONCLUSIONS The most damage develops between 15 minutes and 1 hour and again between 2 and 6 hours after ASDH. The time course of lesion growth supports the approach of early surgery for patients. It furthermore constitutes a basis for further ASDH research with more clearly defined time windows for therapy in animal models.
Collapse
Affiliation(s)
- Daniel Jussen
- Department of Neurosurgery, Goethe University, Frankfurt, Germany; Institute for Neurosurgical Pathophysiology, Johannes Gutenberg University, Mainz, Germany.
| | - Elena Amoruso
- Institute for Neurosurgical Pathophysiology, Johannes Gutenberg University, Mainz, Germany
| | - Oliver Kempski
- Institute for Neurosurgical Pathophysiology, Johannes Gutenberg University, Mainz, Germany
| | - Kristin Lucia
- Department of Neurosurgery, Goethe University, Frankfurt, Germany
| | - Marcus Czabanka
- Department of Neurosurgery, Goethe University, Frankfurt, Germany
| | - Florian Ringel
- Department of Neurosurgery, University Medical Center Mainz, Mainz, Germany
| | - Beat Alessandri
- Institute for Neurosurgical Pathophysiology, Johannes Gutenberg University, Mainz, Germany; Department of Neurosurgery, University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
11
|
Hennigan K, Lavik E. Nature vs. Manmade: Comparing Exosomes and Liposomes for Traumatic Brain Injury. AAPS J 2023; 25:83. [PMID: 37610471 DOI: 10.1208/s12248-023-00849-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 08/08/2023] [Indexed: 08/24/2023] Open
Abstract
Traumatic brain injury (TBI) of all severities is a significant public health burden, causing a range of effects that can lead to death or a diminished quality of life. Liposomes and mesenchymal stem cell-derived exosomes are two drug delivery agents with potential to be leveraged in the treatment of TBI by increasing the efficacy of drug therapies as well as having additional therapeutic effects. They exhibit several physical similarities, but key differences affect their performances as nanocarriers. Liposomes can be produced commercially at scale, and liposomes achieve higher encapsulation efficiency. Meanwhile, the intrinsic cargo and targeting moieties of exosomes, which liposomes lack, give exosomes a greater ability to facilitate neural regeneration, and exosomes do not trigger the infusion reactions that liposomes can. However, there are concerns about both exosomes and liposomes regarding interactions with tumors. The same routes of administration can be used for both exosomes and liposomes, resulting in somewhat different distribution throughout the body. While the effect of the nanocarrier type on accumulation in the brain is not concrete, targeting leads to increased accumulation of both exosomes and liposomes in the brain, upon which on-demand release can be used for both drug deliverers. Although neither have been applied to TBI in humans, preclinical trials have shown their immense potential, as have clinical trials pertaining to other brain injuries and conditions. While questions remain, research thus far shows that the various differences make exosomes a better choice of nanocarrier for TBI.
Collapse
Affiliation(s)
- Kate Hennigan
- Marriotts Ridge High School, Ellicott City, Maryland, 21042, USA
| | - Erin Lavik
- University of Maryland, Baltimore County, 1000 Hilltop Circle, Baltimore, Maryland, 21250, USA.
| |
Collapse
|
12
|
Mansour NO, Elnaem MH, Abdelaziz DH, Barakat M, Dehele IS, Elrggal ME, Abdallah MS. Effects of early adjunctive pharmacotherapy on serum levels of brain injury biomarkers in patients with traumatic brain injury: a systematic review of randomized controlled studies. Front Pharmacol 2023; 14:1185277. [PMID: 37214454 PMCID: PMC10196026 DOI: 10.3389/fphar.2023.1185277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/14/2023] [Indexed: 05/24/2023] Open
Abstract
Objectives: Traumatic brain injury (TBI) is one of the top causes of morbidity and mortality worldwide. The review aimed to discuss and summarize the current evidence on the effectiveness of adjuvant neuroprotective treatments in terms of their effect on brain injury biomarkers in TBI patients. Methods: To identify relevant studies, four scholarly databases, including PubMed, Cochrane, Scopus, and Google Scholar, were systematically searched using predefined search terms. English-language randomized controlled clinical trials reporting changes in brain injury biomarkers, namely, neuron-specific enolase (NSE), glial fibrillary acid protein (GFAP), ubiquitin carboxyl-terminal esterase L1 (UCHL1) and/or S100 beta (S100 ß), were included. The methodological quality of the included studies was assessed using the Cochrane risk-of-bias tool. Results: A total of eleven studies with eight different therapeutic options were investigated; of them, tetracyclines, metformin, and memantine were discovered to be promising choices that could improve neurological outcomes in TBI patients. The most utilized serum biomarkers were NSE and S100 ß followed by GFAP, while none of the included studies quantified UCHL1. The heterogeneity in injury severity categories and measurement timing may affect the overall evaluation of the clinical efficacy of potential therapies. Therefore, unified measurement protocols are highly warranted to inform clinical decisions. Conclusion: Few therapeutic options showed promising results as an adjuvant to standard care in patients with TBI. Several considerations for future work must be directed towards standardizing monitoring biomarkers. Investigating the pharmacotherapy effectiveness using a multimodal biomarker panel is needed. Finally, employing stratified randomization in future clinical trials concerning potential confounders, including age, trauma severity levels, and type, is crucial to inform clinical decisions. Clinical Trial Registration: [https://www.crd.york.ac.uk/prospero/dis], identifier [CRD42022316327].
Collapse
Affiliation(s)
- Noha O. Mansour
- Clinical Pharmacy and Pharmacy Practice Department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Mohamed Hassan Elnaem
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Malaysia
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, United Kingdom
| | - Doaa H. Abdelaziz
- Pharmacy Practice and Clinical Pharmacy Department, Faculty of Pharmacy, Future University in Egypt, Cairo, Egypt
| | - Muna Barakat
- Department of Clinical Pharmacy and Therapeutics, Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
- MEU Research Unit, Middle East University, Amman, Jordan
| | | | | | - Mahmoud S. Abdallah
- Department of Clinical Pharmacy, Faculty of Pharmacy, University of Sadat City, Sadat City, Egypt
| |
Collapse
|
13
|
Lecca D, Hsueh SC, Luo W, Tweedie D, Kim DS, Baig AM, Vargesson N, Kim YK, Hwang I, Kim S, Hoffer BJ, Chiang YH, Greig NH. Novel, thalidomide-like, non-cereblon binding drug tetrafluorobornylphthalimide mitigates inflammation and brain injury. J Biomed Sci 2023; 30:16. [PMID: 36872339 PMCID: PMC9987061 DOI: 10.1186/s12929-023-00907-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/09/2023] [Indexed: 03/07/2023] Open
Abstract
BACKGROUND Quelling microglial-induced excessive neuroinflammation is a potential treatment strategy across neurological disorders, including traumatic brain injury (TBI), and can be achieved by thalidomide-like drugs albeit this approved drug class is compromised by potential teratogenicity. Tetrafluorobornylphthalimide (TFBP) and tetrafluoronorbornylphthalimide (TFNBP) were generated to retain the core phthalimide structure of thalidomide immunomodulatory imide drug (IMiD) class. However, the classical glutarimide ring was replaced by a bridged ring structure. TFBP/TFNBP were hence designed to retain beneficial anti-inflammatory properties of IMiDs but, importantly, hinder cereblon binding that underlies the adverse action of thalidomide-like drugs. METHODS TFBP/TFNBP were synthesized and evaluated for cereblon binding and anti-inflammatory actions in human and rodent cell cultures. Teratogenic potential was assessed in chicken embryos, and in vivo anti-inflammatory actions in rodents challenged with either lipopolysaccharide (LPS) or controlled cortical impact (CCI) moderate traumatic brain injury (TBI). Molecular modeling was performed to provide insight into drug/cereblon binding interactions. RESULTS TFBP/TFNBP reduced markers of inflammation in mouse macrophage-like RAW264.7 cell cultures and in rodents challenged with LPS, lowering proinflammatory cytokines. Binding studies demonstrated minimal interaction with cereblon, with no resulting degradation of teratogenicity-associated transcription factor SALL4 or of teratogenicity in chicken embryo assays. To evaluate the biological relevance of its anti-inflammatory actions, two doses of TFBP were administered to mice at 1 and 24 h post-injury following CCI TBI. Compared to vehicle treatment, TFBP reduced TBI lesion size together with TBI-induction of an activated microglial phenotype, as evaluated by immunohistochemistry 2-weeks post-injury. Behavioral evaluations at 1- and 2-weeks post-injury demonstrated TFBP provided more rapid recovery of TBI-induced motor coordination and balance impairments, versus vehicle treated mice. CONCLUSION TFBP and TFNBP represent a new class of thalidomide-like IMiDs that lower proinflammatory cytokine generation but lack binding to cereblon, the main teratogenicity-associated mechanism. This aspect makes TFBP and TFNBP potentially safer than classic IMiDs for clinical use. TFBP provides a strategy to mitigate excessive neuroinflammation associated with moderate severity TBI to, thereby, improve behavioral outcome measures and warrants further investigation in neurological disorders involving a neuroinflammatory component.
Collapse
Affiliation(s)
- Daniela Lecca
- Drug Design and Development Section, Translational Gerontology Branch, Intramural Research Program National Institute On Aging, NIH, Baltimore, MD, 21224, USA
| | - Shih-Chang Hsueh
- Drug Design and Development Section, Translational Gerontology Branch, Intramural Research Program National Institute On Aging, NIH, Baltimore, MD, 21224, USA
| | - Weiming Luo
- Drug Design and Development Section, Translational Gerontology Branch, Intramural Research Program National Institute On Aging, NIH, Baltimore, MD, 21224, USA
| | - David Tweedie
- Drug Design and Development Section, Translational Gerontology Branch, Intramural Research Program National Institute On Aging, NIH, Baltimore, MD, 21224, USA
| | - Dong Seok Kim
- Aevisbio Inc., Gaithersburg, MD, 20878, USA
- Aevis Bio Inc., Daejeon, 34141, Republic of Korea
| | - Abdul Mannan Baig
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, 74800, Pakistan
| | - Neil Vargesson
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | - Yu Kyung Kim
- Aevis Bio Inc., Daejeon, 34141, Republic of Korea
| | - Inho Hwang
- Aevis Bio Inc., Daejeon, 34141, Republic of Korea
| | - Sun Kim
- Aevis Bio Inc., Daejeon, 34141, Republic of Korea
| | - Barry J Hoffer
- Department of Neurological Surgery, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Yung-Hsiao Chiang
- Neuroscience Research Center, Taipei Medical University, Taipei, 110, Taiwan.
- Department of Neurosurgery, Taipei Medical University Hospital, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.
| | - Nigel H Greig
- Drug Design and Development Section, Translational Gerontology Branch, Intramural Research Program National Institute On Aging, NIH, Baltimore, MD, 21224, USA.
| |
Collapse
|
14
|
Hegdekar N, Sarkar C, Bustos S, Ritzel RM, Hanscom M, Ravishankar P, Philkana D, Wu J, Loane DJ, Lipinski MM. Inhibition of autophagy in microglia and macrophages exacerbates innate immune responses and worsens brain injury outcomes. Autophagy 2023:1-19. [PMID: 36652438 DOI: 10.1080/15548627.2023.2167689] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Excessive and prolonged neuroinflammation following traumatic brain injury (TBI) contributes to long-term tissue damage and poor functional outcomes. However, the mechanisms contributing to exacerbated inflammatory responses after brain injury remain poorly understood. Our previous work showed that macroautophagy/autophagy flux is inhibited in neurons following TBI in mice and contributes to neuronal cell death. In the present study, we demonstrate that autophagy is also inhibited in activated microglia and infiltrating macrophages, and that this potentiates injury-induced neuroinflammatory responses. Macrophage/microglia-specific knockout of the essential autophagy gene Becn1 led to overall increase in neuroinflammation after TBI. In particular, we observed excessive activation of the innate immune responses, including both the type-I interferon and inflammasome pathways. Defects in microglial and macrophage autophagy following injury were associated with decreased phagocytic clearance of danger/damage-associated molecular patterns (DAMP) responsible for activation of the cellular innate immune responses. Our data also demonstrated a role for precision autophagy in targeting and degradation of innate immune pathways components, such as the NLRP3 inflammasome. Finally, inhibition of microglial/macrophage autophagy led to increased neurodegeneration and worse long-term cognitive outcomes after TBI. Conversely, increasing autophagy by treatment with rapamycin decreased inflammation and improved outcomes in wild-type mice after TBI. Overall, our work demonstrates that inhibition of autophagy in microglia and infiltrating macrophages contributes to excessive neuroinflammation following brain injury and in the long term may prevent resolution of inflammation and tissue regeneration.Abbreviations: Becn1/BECN1, beclin 1, autophagy related; CCI, controlled cortical impact; Cybb/CYBB/NOX2: cytochrome b-245, beta polypeptide; DAMP, danger/damage-associated molecular patterns; Il1b/IL1B/Il-1β, interleukin 1 beta; LAP, LC3-associated phagocytosis; Map1lc3b/MAP1LC3/LC3, microtubule-associated protein 1 light chain 3 beta; Mefv/MEFV/TRIM20: Mediterranean fever; Nos2/NOS2/iNOS: nitric oxide synthase 2, inducible; Nlrp3/NLRP3, NLR family, pyrin domain containing 3; Sqstm1/SQSTM1/p62, sequestosome 1; TBI, traumatic brain injury; Tnf/TNF/TNF-α, tumor necrosis factor; Ulk1/ULK1, unc-51 like kinase 1.
Collapse
Affiliation(s)
- Nivedita Hegdekar
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Chinmoy Sarkar
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sabrina Bustos
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rodney M Ritzel
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Neurology, McGovern Medical School, University of Texas, Houston, Tx, USA
| | - Marie Hanscom
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Prarthana Ravishankar
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Deepika Philkana
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Junfang Wu
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - David J Loane
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA.,School of Biochemistry and Immunology, Trinity College, Dublin, Ireland
| | - Marta M Lipinski
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
15
|
Wang X, Li X, Ma L, Chen H, You C. Pharmacological components with neuroprotective effects in the management of traumatic brain injury: evidence from network meta-analysis. Neurol Sci 2023; 44:1665-1678. [PMID: 36642741 DOI: 10.1007/s10072-023-06600-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 12/31/2022] [Indexed: 01/17/2023]
Abstract
BACKGROUND Neuroprotective drugs have been used to prevent secondary brain injury in patients with traumatic brain injury; however, the optimal medication remains questionable. We performed a Bayesian network meta-analysis to evaluate the safety and efficacy of different medications with known neuroprotective properties in this group of patients. METHODS Several databases were searched to identify any eligible trials comparing pharmacological components with confirmed neuroprotective mechanisms. Bayesian network meta-analysis was performed to combine direct and indirect evidence. The surface under the cumulative ranking curve was obtained to determine the ranking probability of the treatment agents for each outcome. The primary outcome was all-cause mortality. RESULTS A total of 23 trials comprising 4,325 participants were identified. The pooled relative risk (RR) showed administration of erythropoietin (RR: 0.68; 95% CrI: 0.50-0.93) and propranolol (RR: 0.43; 95% CrI: 0.20-0.85) decreased all-cause mortality compared with placebo. We also found erythropoietin (RR: 1.55; 95% CrI: 1.03-2.35), propranolol (RR: 1.52; 95% CrI: 1.05-2.20), and progesterone (RR: 1.47; 95% CrI: 1.03-2.10) showed better efficacy in functional recovery. CONCLUSION Overall, erythropoietin and propranolol were associated with reduced mortality in adults with traumatic brain injury. These treatment agents were also associated with improved functional outcomes.
Collapse
Affiliation(s)
- Xing Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Xiaolong Li
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Lu Ma
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Hui Chen
- Department of Neurosurgery, Sichuan Friendship Hospital, Chengdu, Sichuan, People's Republic of China.
| | - Chao You
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.
- West China Brain Research Centre, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
16
|
Huang J, Zhu H, Yu P, Ma Y, Gong J, Fu Y, Song H, Huang M, Luo J, Jiang J, Gao X, Feng J, Jiang G. Recombinant High-Density Lipoprotein Boosts the Therapeutic Efficacy of Mild Hypothermia in Traumatic Brain Injury. ACS APPLIED MATERIALS & INTERFACES 2023; 15:26-38. [PMID: 35833835 DOI: 10.1021/acsami.2c02940] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Traumatic brain injury (TBI) leads to neuropsychiatric symptoms and increased risk of neurodegenerative disorders. Mild hypothermia is commonly used in patients suffering from severe TBI. However, its effect for long-term protection is limited, mostly because of its insufficient anti-inflammatory and neuroprotective efficacy and restricted treatment duration. Recombinant high-density lipoprotein (rHDL), which possesses anti-inflammatory and antioxidant activity and blood-brain barrier (BBB) permeability, was expected to potentially strengthen the therapeutic effect of mild hypothermia in TBI treatment. To test this hypothesis and optimize the regimen for combination therapy, the efficacy of mild hypothermia plus concurrent or sequential rHDL on oxidative stress, inflammatory reaction, and cell survival in the damaged brain cells was evaluated. It was found that the effect of combining mild hypothermia with concurrent rHDL was modest, as mild hypothermia inhibited the cellular uptake and lesion-site-targeting delivery of rHDL. In contrast, the combination of mild hypothermia with sequential rHDL more powerfully improved the anti-inflammatory and antioxidant activities, promoted nerve cell survival and BBB restoration, and ameliorated neurologic changes, which thus remarkably restored the spatial learning and memory ability of TBI mice. Collectively, these findings suggest that rHDL may serve as a novel nanomedicine for adjunctive therapy of TBI and highlight the importance of timing of combination therapy for optimal treatment outcome.
Collapse
Affiliation(s)
- Jialin Huang
- Brain Injury Center, Renji Hospital, Shanghai Institute of Head Trauma, School of Medicine, Shanghai Jiao Tong University, No. 160 Pujian Road, Shanghai 200127, China
| | - Han Zhu
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, No. 2800 Gongwei Road, Shanghai 201399, China
| | - Ping Yu
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197 Rui Jin Er Road, Shanghai 200025, China
| | - Yuxiao Ma
- Brain Injury Center, Renji Hospital, Shanghai Institute of Head Trauma, School of Medicine, Shanghai Jiao Tong University, No. 160 Pujian Road, Shanghai 200127, China
| | - Jingru Gong
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, No. 2800 Gongwei Road, Shanghai 201399, China
| | - Yuli Fu
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Huahua Song
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Meng Huang
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Jing Luo
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Jiyao Jiang
- Brain Injury Center, Renji Hospital, Shanghai Institute of Head Trauma, School of Medicine, Shanghai Jiao Tong University, No. 160 Pujian Road, Shanghai 200127, China
| | - Xiaoling Gao
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Junfeng Feng
- Brain Injury Center, Renji Hospital, Shanghai Institute of Head Trauma, School of Medicine, Shanghai Jiao Tong University, No. 160 Pujian Road, Shanghai 200127, China
| | - Gan Jiang
- Department of Pharmacology and Chemical Biology, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| |
Collapse
|
17
|
Rauchman SH, Zubair A, Jacob B, Rauchman D, Pinkhasov A, Placantonakis DG, Reiss AB. Traumatic brain injury: Mechanisms, manifestations, and visual sequelae. Front Neurosci 2023; 17:1090672. [PMID: 36908792 PMCID: PMC9995859 DOI: 10.3389/fnins.2023.1090672] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Traumatic brain injury (TBI) results when external physical forces impact the head with sufficient intensity to cause damage to the brain. TBI can be mild, moderate, or severe and may have long-term consequences including visual difficulties, cognitive deficits, headache, pain, sleep disturbances, and post-traumatic epilepsy. Disruption of the normal functioning of the brain leads to a cascade of effects with molecular and anatomical changes, persistent neuronal hyperexcitation, neuroinflammation, and neuronal loss. Destructive processes that occur at the cellular and molecular level lead to inflammation, oxidative stress, calcium dysregulation, and apoptosis. Vascular damage, ischemia and loss of blood brain barrier integrity contribute to destruction of brain tissue. This review focuses on the cellular damage incited during TBI and the frequently life-altering lasting effects of this destruction on vision, cognition, balance, and sleep. The wide range of visual complaints associated with TBI are addressed and repair processes where there is potential for intervention and neuronal preservation are highlighted.
Collapse
Affiliation(s)
| | - Aarij Zubair
- NYU Long Island School of Medicine, Mineola, NY, United States
| | - Benna Jacob
- NYU Long Island School of Medicine, Mineola, NY, United States
| | - Danielle Rauchman
- Department of Neuroscience, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Aaron Pinkhasov
- NYU Long Island School of Medicine, Mineola, NY, United States
| | | | - Allison B Reiss
- NYU Long Island School of Medicine, Mineola, NY, United States
| |
Collapse
|
18
|
Drieu A, Lanquetin A, Prunotto P, Gulhan Z, Pédron S, Vegliante G, Tolomeo D, Serrière S, Vercouillie J, Galineau L, Tauber C, Kuhnast B, Rubio M, Zanier ER, Levard D, Chalon S, Vivien D, Ali C. Persistent neuroinflammation and behavioural deficits after single mild traumatic brain injury. J Cereb Blood Flow Metab 2022; 42:2216-2229. [PMID: 35945692 PMCID: PMC9670002 DOI: 10.1177/0271678x221119288] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 12/14/2022]
Abstract
Despite an apparently silent imaging, some patients with mild traumatic brain injury (TBI) experience cognitive dysfunctions, which may persist chronically. Brain changes responsible for these dysfunctions are unclear and commonly overlooked. It is thus crucial to increase our understanding of the mechanisms linking the initial event to the functional deficits, and to provide objective evidence of brain tissue alterations underpinning these deficits. We first set up a murine model of closed-head controlled cortical impact, which provoked persistent cognitive and sensorimotor deficits, despite no evidence of brain contusion or bleeding on MRI, thus recapitulating features of mild TBI. Molecular MRI for P-selectin, a key adhesion molecule, detected no sign of cerebrovascular inflammation after mild TBI, as confirmed by immunostainings. By contrast, in vivo PET imaging with the TSPO ligand [18F]DPA-714 demonstrated persisting signs of neuroinflammation in the ipsilateral cortex and hippocampus after mild TBI. Interestingly, immunohistochemical analyses confirmed these spatio-temporal profiles, showing a robust parenchymal astrogliosis and microgliosis, at least up to 3 weeks post-injury in both the cortex and hippocampus. In conclusion, we show that even one single mild TBI induces long-term behavioural deficits, associated with a persistent neuro-inflammatory status that can be detected by PET imaging.
Collapse
Affiliation(s)
- Antoine Drieu
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237,
Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain
@ Caen-Normandie, Cyceron, France
| | - Anastasia Lanquetin
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237,
Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain
@ Caen-Normandie, Cyceron, France
| | - Paul Prunotto
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237,
Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain
@ Caen-Normandie, Cyceron, France
| | - Zuhal Gulhan
- UMR 1253, iBrain, Université de Tours, INSERM, Tours,
France
| | - Swannie Pédron
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237,
Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain
@ Caen-Normandie, Cyceron, France
| | - Gloria Vegliante
- Department of Neuroscience, Istituto di Ricerche Farmacologiche
Mario Negri, IRCCS, Milan, Italy
| | - Daniele Tolomeo
- Department of Neuroscience, Istituto di Ricerche Farmacologiche
Mario Negri, IRCCS, Milan, Italy
| | - Sophie Serrière
- UMR 1253, iBrain, Université de Tours, INSERM, Tours,
France
| | | | | | - Clovis Tauber
- UMR 1253, iBrain, Université de Tours, INSERM, Tours,
France
| | - Bertrand Kuhnast
- IMIV, Service Hospitalier Frédéric Joliot, CEA, Inserm,
Université Paris Sud, CNRS, Université Paris-Saclay, Orsay, France
| | - Marina Rubio
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237,
Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain
@ Caen-Normandie, Cyceron, France
| | - Elisa R Zanier
- Department of Neuroscience, Istituto di Ricerche Farmacologiche
Mario Negri, IRCCS, Milan, Italy
| | - Damien Levard
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237,
Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain
@ Caen-Normandie, Cyceron, France
| | - Sylvie Chalon
- UMR 1253, iBrain, Université de Tours, INSERM, Tours,
France
| | - Denis Vivien
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237,
Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain
@ Caen-Normandie, Cyceron, France
- Department of Clinical Research, Caen-Normandie Hospital (CHU),
Caen, France
| | - Carine Ali
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237,
Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain
@ Caen-Normandie, Cyceron, France
| |
Collapse
|
19
|
Nasre-Nasser RG, Severo MMR, Pires GN, Hort MA, Arbo BD. Effects of Progesterone on Preclinical Animal Models of Traumatic Brain Injury: Systematic Review and Meta-analysis. Mol Neurobiol 2022; 59:6341-6362. [PMID: 35922729 DOI: 10.1007/s12035-022-02970-9] [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: 04/05/2022] [Accepted: 07/21/2022] [Indexed: 12/09/2022]
Abstract
Since the publication of two phase III clinical trials not supporting the use of progesterone in patients with traumatic brain injury (TBI), several possible explanations have been postulated, including limitations in the analysis of results from preclinical evidence. Therefore, to address this question, a systematic review and meta-analysis was performed to evaluate the effects of progesterone as a neuroprotective agent in preclinical animal models of TBI. A total of 48 studies were included for review: 29 evaluated brain edema, 21 evaluated lesion size, and 0 studies reported the survival rate. In the meta-analysis, it was found that progesterone reduced brain edema (effect size - 1.73 [- 2.02, - 1.44], p < 0.0001) and lesion volume (effect size - 0.40 [- 0.65, - 0.14], p = 0.002). Lack of details in the studies hindered the assessment of risk of bias (through the SYRCLE tool). A funnel plot asymmetry was detected, suggesting a possible publication bias. In conclusion, preclinical studies show that progesterone has an anti-edema effect in animal models of TBI, decreasing lesion volume or increasing remaining tissue. However, more studies are needed using assessing methods with lower risk of histological artifacts.
Collapse
Affiliation(s)
- Raif Gregorio Nasre-Nasser
- Programa de Pós-Graduação Em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal Do Rio Grande (FURG), Porto Alegre, Rio Grande do Sul, Brazil
| | - Maria Manoela Rezende Severo
- Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul (UFRGS), Rua Ramiro Barcelos 2600, Building UFRGS 21116, Room 430, Zip code, Porto Alegre - RS, 90035-003, Brazil
| | - Gabriel Natan Pires
- Departamento de Psicobiologia, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- Brazilian Reproducibility Initiative in Preclinical Systematic Review and Meta-Analysis (BRISA), Rio de Janeiro, Brazil
| | - Mariana Appel Hort
- Programa de Pós-Graduação Em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal Do Rio Grande (FURG), Porto Alegre, Rio Grande do Sul, Brazil
| | - Bruno Dutra Arbo
- Programa de Pós-Graduação Em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal Do Rio Grande (FURG), Porto Alegre, Rio Grande do Sul, Brazil.
- Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul (UFRGS), Rua Ramiro Barcelos 2600, Building UFRGS 21116, Room 430, Zip code, Porto Alegre - RS, 90035-003, Brazil.
| |
Collapse
|
20
|
Lee D, Tomita Y, Miwa Y, Shinojima A, Ban N, Yamaguchi S, Nishioka K, Negishi K, Yoshino J, Kurihara T. Nicotinamide Mononucleotide Prevents Retinal Dysfunction in a Mouse Model of Retinal Ischemia/Reperfusion Injury. Int J Mol Sci 2022; 23:ijms231911228. [PMID: 36232528 PMCID: PMC9570481 DOI: 10.3390/ijms231911228] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Retinal ischemia/reperfusion (I/R) injury can cause severe vision impairment. Retinal I/R injury is associated with pathological increases in reactive oxygen species and inflammation, resulting in retinal neuronal cell death. To date, effective therapies have not been developed. Nicotinamide mononucleotide (NMN), a key nicotinamide adenine dinucleotide (NAD+) intermediate, has been shown to exert neuroprotection for retinal diseases. However, it remains unclear whether NMN can prevent retinal I/R injury. Thus, we aimed to determine whether NMN therapy is useful for retinal I/R injury-induced retinal degeneration. One day after NMN intraperitoneal (IP) injection, adult mice were subjected to retinal I/R injury. Then, the mice were injected with NMN once every day for three days. Electroretinography and immunohistochemistry were used to measure retinal functional alterations and retinal inflammation, respectively. The protective effect of NMN administration was further examined using a retinal cell line, 661W, under CoCl2-induced oxidative stress conditions. NMN IP injection significantly suppressed retinal functional damage, as well as inflammation. NMN treatment showed protective effects against oxidative stress-induced cell death. The antioxidant pathway (Nrf2 and Hmox-1) was activated by NMN treatment. In conclusion, NMN could be a promising preventive neuroprotective drug for ischemic retinopathy.
Collapse
Affiliation(s)
- Deokho Lee
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yohei Tomita
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yukihiro Miwa
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Aichi Animal Eye Clinic, Nagoya 466-0827, Japan
| | - Ari Shinojima
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Norimitsu Ban
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shintaro Yamaguchi
- Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Ken Nishioka
- Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Jun Yoshino
- Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Toshihide Kurihara
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
- Correspondence:
| |
Collapse
|
21
|
Kalra S, Malik R, Singh G, Bhatia S, Al-Harrasi A, Mohan S, Albratty M, Albarrati A, Tambuwala MM. Pathogenesis and management of traumatic brain injury (TBI): role of neuroinflammation and anti-inflammatory drugs. Inflammopharmacology 2022; 30:1153-1166. [PMID: 35802283 PMCID: PMC9293826 DOI: 10.1007/s10787-022-01017-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/06/2022] [Indexed: 02/08/2023]
Abstract
Traumatic brain injury (TBI) is an important global health concern that represents a leading cause of death and disability. It occurs due to direct impact or hit on the head caused by factors such as motor vehicles, crushes, and assaults. During the past decade, an abundance of new evidence highlighted the importance of inflammation in the secondary damage response that contributes to neurodegenerative and neurological deficits after TBI. It results in disruption of the blood-brain barrier (BBB) and initiates the release of macrophages, neutrophils, and lymphocytes at the injury site. A growing number of researchers have discovered various signalling pathways associated with the initiation and progression of inflammation. Targeting different signalling pathways (NF-κB, JAK/STAT, MAPKs, PI3K/Akt/mTOR, GSK-3, Nrf2, RhoGTPase, TGF-β1, and NLRP3) helps in the development of novel anti-inflammatory drugs in the management of TBI. Several synthetic and herbal drugs with both anti-inflammatory and neuroprotective potential showed effective results. This review summarizes different signalling pathways, associated pathologies, inflammatory mediators, pharmacological potential, current status, and challenges with anti-inflammatory drugs.
Collapse
Affiliation(s)
- Sunishtha Kalra
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Rohit Malik
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Govind Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India.
| | - Saurabh Bhatia
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India. .,Natural and Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, Nizwa, Oman.
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz, Nizwa, Oman
| | - Syam Mohan
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India.,Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Ali Albarrati
- Rehabilitation Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Northern Ireland, UK.
| |
Collapse
|
22
|
Herr SA, Gardeen SS, Low PS, Shi R. Targeted delivery of acrolein scavenger hydralazine in spinal cord injury using folate-linker-drug conjugation. Free Radic Biol Med 2022; 184:66-73. [PMID: 35398493 DOI: 10.1016/j.freeradbiomed.2022.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/30/2022] [Accepted: 04/03/2022] [Indexed: 11/26/2022]
Abstract
Oxidative stress has been shown to play a critical pathogenic role in functional loss after spinal cord injury (SCI). As a direct result of oxidative stress, lipid peroxidation-derived aldehydes have emerged as key culprits that sustain secondary injury and contribute significantly to pathological outcomes. Acrolein, a neurotoxin, has been shown to be elevated in SCI and can result in post-SCI neurological deficits. Reducing acrolein has therefore emerged as a novel and effective therapeutic strategy in SCI. Previous studies have revealed that hydralazine, an FDA approved blood pressure lowering medication, when administered after SCI shows strong acrolein scavenging capabilities and significantly improves cellular and behavioral outcomes. However, while effective at scavenging acrolein, hydralazine's blood pressure lowering activity can have a detrimental impact on neurotrauma patients. Here, our goal was to preserve the acrolein scavenging capability while mitigating the effect of hydralazine on blood pressure. We accomplished this using a folate-targeted delivery system to deploy hydralazine to the folate receptor positive inflammatory site of the cord injury. Using a model of rat SCI, we found that this system is effective for targeting the injury site, and that folate targeted hydralazine can scavenge acrolein without significantly impacting blood pressure.
Collapse
Affiliation(s)
- Seth A Herr
- Center for Paralysis Research & Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University. Lynn Hall, 625 N Harrison St. West Lafayette, IN, 47907, USA.
| | - Spencer S Gardeen
- Department of Chemistry, College of Science, Purdue University. Drug Discovery Building, 720 Clinic Dr. West Lafayette, IN, 47907, USA.
| | - Philip S Low
- Department of Chemistry, College of Science, Purdue University. Drug Discovery Building, 720 Clinic Dr. West Lafayette, IN, 47907, USA.
| | - Riyi Shi
- Center for Paralysis Research & Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University. Lynn Hall, 625 N Harrison St. West Lafayette, IN, 47907, USA.
| |
Collapse
|
23
|
Ribonuclease-1 treatment after traumatic brain injury preserves blood-brain barrier integrity and delays secondary brain damage in mice. Sci Rep 2022; 12:5731. [PMID: 35388024 PMCID: PMC8986812 DOI: 10.1038/s41598-022-09326-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 03/22/2022] [Indexed: 11/08/2022] Open
Abstract
Traumatic brain injury (TBI) involves primary mechanical damage and delayed secondary damage caused by vascular dysfunction and neuroinflammation. Intracellular components released into the parenchyma and systemic circulation, termed danger-associated molecular patterns (DAMPs), are major drivers of vascular dysfunction and neuroinflammation. These DAMPs include cell-free RNAs (cfRNAs), which damage the blood-brain barrier (BBB), thereby promoting edema, procoagulatory processes, and infiltration of inflammatory cells. We tested the hypothesis that intraperitoneal injection of Ribonuclease-1 (RNase1, two doses of 20, 60, or 180 µg/kg) at 30 min and 12 h after controlled-cortical-impact (CCI) can reduce secondary lesion expansion compared to vehicle treatment 24 h and 120 h post-CCI. The lowest total dose (40 µg/kg) was most effective at reducing lesion volume (- 31% RNase 40 µg/kg vs. vehicle), brain water accumulation (- 5.5%), and loss of BBB integrity (- 21.6%) at 24 h post-CCI. RNase1 also reduced perilesional leukocyte recruitment (- 53.3%) and microglial activation (- 18.3%) at 120 h post-CCI, but there was no difference in lesion volume at this time and no functional benefit. Treatment with RNase1 in the early phase following TBI stabilizes the BBB and impedes leukocyte immigration, thereby suppressing neuroinflammation. RNase1-treatment may be a novel approach to delay brain injury to extend the window for treatment opportunities after TBI.
Collapse
|
24
|
Role of Inflammation in Traumatic Brain Injury-Associated Risk for Neuropsychiatric Disorders: State of the Evidence and Where Do We Go From Here. Biol Psychiatry 2022; 91:438-448. [PMID: 34955170 DOI: 10.1016/j.biopsych.2021.11.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/01/2021] [Accepted: 11/02/2021] [Indexed: 02/06/2023]
Abstract
In the past decade, there has been an increasing awareness that traumatic brain injury (TBI) and concussion substantially increase the risk for developing psychiatric disorders. Even mild TBI increases the risk for depression and anxiety disorders such as posttraumatic stress disorder by two- to threefold, predisposing patients to further functional impairment. This strong epidemiological link supports examination of potential mechanisms driving neuropsychiatric symptom development after TBI. One potential mechanism for increased neuropsychiatric symptoms after TBI is via inflammatory processes, as central nervous system inflammation can last years after initial injury. There is emerging preliminary evidence that TBI patients with posttraumatic stress disorder or depression exhibit increased central and peripheral inflammatory markers compared with TBI patients without these comorbidities. Growing evidence has demonstrated that immune signaling in animals plays an integral role in depressive- and anxiety-like behaviors after severe stress or brain injury. In this review, we will 1) discuss current evidence for chronic inflammation after TBI in the development of neuropsychiatric symptoms, 2) highlight potential microglial activation and cytokine signaling contributions, and 3) discuss potential promise and pitfalls for immune-targeted interventions and biomarker strategies to identify and treat TBI patients with immune-related neuropsychiatric symptoms.
Collapse
|
25
|
Sayeed I, Stein DG. Response to Korley et al.: Progesterone Treatment Does Not Decrease Serum Levels of Biomarkers of Glial and Neuronal Cell Injury in Moderate and Severe TBI Subjects: A Secondary Analysis of the Progesterone for Traumatic Brain Injury, Experimental Clinical Treatment (ProTECT) III Trial (DOI: 10.1089/neu.2020.7072). J Neurotrauma 2021; 38:2923-2926. [PMID: 34130480 DOI: 10.1089/neu.2021.0064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Affiliation(s)
- Iqbal Sayeed
- Department of Emergency Medicine, Emory University, Atlanta, Georgia, USA
| | - Donald G Stein
- Department of Emergency Medicine, Emory University, Atlanta, Georgia, USA
| |
Collapse
|