51
|
King ZA, Sheth KN, Kimberly WT, Simard JM. Profile of intravenous glyburide for the prevention of cerebral edema following large hemispheric infarction: evidence to date. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:2539-2552. [PMID: 30147301 PMCID: PMC6101021 DOI: 10.2147/dddt.s150043] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Glyburide (also known as glibenclamide) is a second-generation sulfonylurea drug that inhibits sulfonylurea receptor 1 (Sur1) at nanomolar concentrations. Long used to target KATP (Sur1–Kir6.2) channels for the treatment of diabetes mellitus type 2, glyburide was recently repurposed to target Sur1–transient receptor potential melastatin 4 (Trpm4) channels in acute central nervous system injury. Discovered nearly two decades ago, SUR1–TRPM4 has emerged as a critical target in stroke, specifically in large hemispheric infarction, which is characterized by edema formation and life-threatening brain swelling. Following ischemia, SUR1–TRPM4 channels are transcriptionally upregulated in all cells of the neurovascular unit, including neurons, astrocytes, microglia, oligodendrocytes and microvascular endothelial cells. Work by several independent laboratories has linked SUR1–TRPM4 to edema formation, with blockade by glyburide reducing brain swelling and death in preclinical models. Recent work showed that, following ischemia, SUR1–TRPM4 co-assembles with aquaporin-4 to mediate cellular swelling of astrocytes, which contributes to brain swelling. Additionally, recent work linked SUR1–TRPM4 to secretion of matrix metalloproteinase-9 (MMP-9) induced by recombinant tissue plasminogen activator in activated brain endothelial cells, with blockade of SUR1–TRPM4 by glyburide reducing MMP-9 and hemorrhagic transformation in preclinical models with recombinant tissue plasminogen activator. The recently completed GAMES (Glyburide Advantage in Malignant Edema and Stroke) clinical trials on patients with large hemispheric infarctions treated with intravenous glyburide (RP-1127) revealed promising findings with regard to brain swelling (midline shift), MMP-9, functional outcomes and mortality. Here, we review key elements of the basic science, preclinical experiments and clinical studies, both retrospective and prospective, on glyburide in focal cerebral ischemia and stroke.
Collapse
Affiliation(s)
- Zachary A King
- Department of Neurology, Division of Neurocritical Care and Emergency Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Kevin N Sheth
- Department of Neurology, Division of Neurocritical Care and Emergency Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - W Taylor Kimberly
- Department of Neurology, Division of Neurocritical Care and Emergency Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA,
| |
Collapse
|
52
|
Pathophysiology and treatment of cerebral edema in traumatic brain injury. Neuropharmacology 2018; 145:230-246. [PMID: 30086289 DOI: 10.1016/j.neuropharm.2018.08.004] [Citation(s) in RCA: 244] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/24/2018] [Accepted: 08/03/2018] [Indexed: 12/30/2022]
Abstract
Cerebral edema (CE) and resultant intracranial hypertension are associated with unfavorable prognosis in traumatic brain injury (TBI). CE is a leading cause of in-hospital mortality, occurring in >60% of patients with mass lesions, and ∼15% of those with normal initial computed tomography scans. After treatment of mass lesions in severe TBI, an important focus of acute neurocritical care is evaluating and managing the secondary injury process of CE and resultant intracranial hypertension. This review focuses on a contemporary understanding of various pathophysiologic pathways contributing to CE, with a subsequent description of potential targeted therapies. There is a discussion of identified cellular/cytotoxic contributors to CE, as well as mechanisms that influence blood-brain-barrier (BBB) disruption/vasogenic edema, with the caveat that this distinction may be somewhat artificial since molecular processes contributing to these pathways are interrelated. While an exhaustive discussion of all pathways with putative contributions to CE is beyond the scope of this review, the roles of some key contributors are highlighted, and references are provided for further details. Potential future molecular targets for treating CE are presented based on pathophysiologic mechanisms. We thus aim to provide a translational synopsis of present and future strategies targeting CE after TBI in the context of a paradigm shift towards precision medicine. This article is part of the Special Issue entitled "Novel Treatments for Traumatic Brain Injury".
Collapse
|
53
|
Zhu S, Gao X, Huang K, Gu Y, Hu Y, Wu Y, Ji Z, Wang Q, Pan S. Glibenclamide Enhances the Therapeutic Benefits of Early Hypothermia after Severe Stroke in Rats. Aging Dis 2018; 9:685-695. [PMID: 30090656 PMCID: PMC6065285 DOI: 10.14336/ad.2017.0927] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 09/27/2017] [Indexed: 01/20/2023] Open
Abstract
Glibenclamide (GBC) is an antidiabetic drug that is in a class of medications known as sulfonylureas, which play critical roles in attenuating brain edema and reducing mortality in ischemic stroke patients. Therapeutic hypothermia (TH) is another robust neuroprotectant that prevents brain swelling and improves the neurological outcomes of stroke patients. However, whether the combination of GBC and TH can be used as a reliable neuroprotectant in ischemic stroke remains largely unknown. We used the middle cerebral artery occlusion (MCAO) rat model as well as oxygen and glucose deprivation-reoxygenation (OGD/R) endothelial cells as ischemic stroke models to investigate the efficacy and mechanisms of treating ischemic stroke with the combination of GBC and TH. The serum glucose, mortality rate, neurobehavioral functions, tight junctions, endothelial cells and inflammatory cytokines were evaluated in the stroke models after treatment with GBC, TH or the combination of them. After 5-hour occlusion and subsequent reperfusion, rats exhibited a large volume of hemispheric swelling and a high mortality rate. Stroke rats treated with the combined therapy did not exhibit hypoglycemia. The combination of GBC and TH exhibited synergistic neuroprotective effects in stroke rats that were associated with greater reductions in edema volume, better improvement in neurobehavioral functions, prevention of tight junction loss, and reduction of expression of the inflammatory cytokines COX-2 and iNOS. In OGD/R endothelia cells, the combination reduced endothelial cell death. This study demonstrated that both GBC and TH are neuroprotective after the severe stroke; however, combined therapy with GBC and TH enhanced the efficiency and efficacy of the effects of TH and GBC in the treatment of ischemia. This combined therapy may facilitate the clinical translation of TH management for severe stroke. The combination of GBC and TH seems to be a feasible and promising clinical strategy to alleviate cerebral injury following severe stroke.
Collapse
Affiliation(s)
- Shuzhen Zhu
- 1Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,2Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoya Gao
- 1Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,2Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Kaibin Huang
- 1Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yong Gu
- 1Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yafang Hu
- 1Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yongming Wu
- 1Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhong Ji
- 1Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qing Wang
- 2Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Suyue Pan
- 1Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
54
|
Irimia A, Van Horn JD, Vespa PM. Cerebral microhemorrhages due to traumatic brain injury and their effects on the aging human brain. Neurobiol Aging 2018; 66:158-164. [PMID: 29579686 PMCID: PMC5924627 DOI: 10.1016/j.neurobiolaging.2018.02.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 02/24/2018] [Accepted: 02/27/2018] [Indexed: 01/08/2023]
Abstract
Although cerebral microbleeds (CMBs) are frequently associated with traumatic brain injury (TBI), their effects on clinical outcome after TBI remain controversial and poorly understood, particularly in older adults. Here we (1) highlight major challenges and opportunities associated with studying the effects of TBI-mediated CMBs; (2) review the evidence on their potential effects on cognitive and neural outcome as a function of age at injury; and (3) suggest priorities for future research on understanding the clinical implications of CMBs. Although TBI-mediated CMBs are likely distinct from those due to cerebral amyloid angiopathy or other neurodegenerative diseases, the effects of these 2 CMB types on brain function may share common features. Furthermore, in older TBI victims, the incidence of TBI-mediated CMBs may approximate that of cerebral amyloid angiopathy-related CMBs, and thus warrants detailed study. Because the alterations effected by CMBs on brain structure and function are both unique and age-dependent, it seems likely that novel, age-tailored therapeutic approaches are necessary for the adequate clinical interpretation and treatment of these ubiquitous and underappreciated TBI sequelae.
Collapse
Affiliation(s)
- Andrei Irimia
- Ethel Percy Andrus Gerontology Center, USC Leonard Davis School of Gerontology, University of Southern California, Los Angeles CA, USA.
| | - John D Van Horn
- USC Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Paul M Vespa
- Departments of Neurosurgery and Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
55
|
Jeffery N, Boudreau CE, Konarik M, Mays T, Fajt V. Pharmacokinetics and safety of oral glyburide in dogs with acute spinal cord injury. PeerJ 2018; 6:e4387. [PMID: 29503767 PMCID: PMC5831157 DOI: 10.7717/peerj.4387] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 01/29/2018] [Indexed: 01/04/2023] Open
Abstract
Background Glyburide (also known as glibenclamide) is effective in reducing the severity of tissue destruction and improving functional outcome after experimental spinal cord injury in rodents and so has promise as a therapy in humans. There are many important differences between spinal cord injury in experimental animals and in human clinical cases, making it difficult to introduce new therapies into clinical practice. Spinal cord injury is also common in pet dogs and requires new effective therapies, meaning that they can act as a translational model for the human condition while also deriving direct benefits from such research. In this study we investigated the pharmacokinetics and safety of glyburide in dogs with clinical spinal cord injury. Methods We recruited dogs that had incurred an acute thoracolumbar spinal cord injury within the previous 72 h. These had become acutely non-ambulatory on the pelvic limbs and were admitted to our veterinary hospitals to undergo anesthesia, cross sectional diagnostic imaging, and surgical decompression. Oral glyburide was given to each dog at a dose of 75 mcg/kg. In five dogs, we measured blood glucose concentrations for 10 h after a single oral dose. In six dogs, we measured serum glyburide and glucose concentrations for 24 h and estimated pharmacokinetic parameters to estimate a suitable dose for use in a subsequent clinical trial in similarly affected dogs. Results No detrimental effects of glyburide administration were detected in any participating dog. Peak serum concentrations of glyburide were attained at a mean of 13 h after dosing, and mean apparent elimination half-life was approximately 7 h. Observed mean maximum plasma concentration was 31 ng/mL. At the glyburide dose administered there was no observable association between glyburide and glucose concentrations in blood. Discussion Our data suggest that glyburide can be safely administered to dogs that are undergoing anesthesia, imaging and surgery for treatment of their acute spinal cord injury and can attain clinically-relevant serum concentrations without developing hazardous hypoglycemia. Serum glyburide concentrations achieved in this study suggest that a loading dose of 150 mcg/kg followed by repeat doses of 75 mcg/kg at 8-hourly intervals would lead to serum glyburide concentrations of 25-50 ng/mL within an acceptably short enough period after oral administration to be appropriate for a clinical trial in canine spinal cord injury.
Collapse
Affiliation(s)
- Nick Jeffery
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States of America
| | - C Elizabeth Boudreau
- Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States of America
| | - Megan Konarik
- Veterinary Medical Diagnostic Laboratory, Texas A&M University, College Station, TX, United States of America
| | - Travis Mays
- Veterinary Medical Diagnostic Laboratory, Texas A&M University, College Station, TX, United States of America
| | - Virginia Fajt
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX, United States of America
| |
Collapse
|
56
|
Bodanapally UK, Shanmuganathan K, Issa G, Dreizin D, Li G, Sudini K, Fleiter TR. Dual-Energy CT in Hemorrhagic Progression of Cerebral Contusion: Overestimation of Hematoma Volumes on Standard 120-kV Images and Rectification with Virtual High-Energy Monochromatic Images after Contrast-Enhanced Whole-Body Imaging. AJNR Am J Neuroradiol 2018; 39:658-662. [PMID: 29439124 DOI: 10.3174/ajnr.a5558] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 12/11/2017] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE In patients with hemorrhagic contusions, hematoma volumes are overestimated on follow-up standard 120-kV images obtained after contrast-enhanced whole-body CT. We aimed to retrospectively determine hemorrhagic progression of contusion rates on 120-kV and 190-keV images derived from dual-energy CT and the magnitude of hematoma volume overestimation. MATERIALS AND METHODS We retrospectively analyzed admission and follow-up CT studies in 40 patients with hemorrhagic contusions. After annotating the contusions, we measured volumes from admission and follow-up 120-kV and 190-keV images using semiautomated 3D segmentation. Bland-Altman analysis was used for hematoma volume comparison. RESULTS On 120-kV images, hemorrhagic progression of contusions was detected in 24 of the 40 patients, while only 17 patients had hemorrhagic progression of contusions on 190-keV images (P = .008). Hematoma volumes were systematically overestimated on follow-up 120-kV images (9.68 versus 8 mm3; mean difference, 1.68 mm3; standard error, 0.37; P < .001) compared with 190-keV images. There was no significant difference in volumes between admission 120-kV and 190-keV images. Mean and median percentages of overestimation were 29% (95% CI, 18-39) and 22% (quartile 3 - quartile 1 = 36.8), respectively. CONCLUSIONS The 120-kV images, which are comparable with single-energy CT images, significantly overestimated the hematoma volumes, hence the rate of hemorrhagic progression of contusions, after contrast-enhanced whole-body CT. Hence, follow-up of hemorrhagic contusions should be performed on dual-energy CT, and 190-keV images should be used for the assessment of hematoma volumes.
Collapse
Affiliation(s)
- U K Bodanapally
- From the Department of Diagnostic Radiology and Nuclear Medicine (U.K.B., K.S., G.I., D.D., G.L., T.R.F.), R Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, Maryland
| | - K Shanmuganathan
- From the Department of Diagnostic Radiology and Nuclear Medicine (U.K.B., K.S., G.I., D.D., G.L., T.R.F.), R Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, Maryland.,Department of Environmental Health Sciences (K.S.), Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - G Issa
- From the Department of Diagnostic Radiology and Nuclear Medicine (U.K.B., K.S., G.I., D.D., G.L., T.R.F.), R Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, Maryland
| | - D Dreizin
- From the Department of Diagnostic Radiology and Nuclear Medicine (U.K.B., K.S., G.I., D.D., G.L., T.R.F.), R Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, Maryland
| | - G Li
- From the Department of Diagnostic Radiology and Nuclear Medicine (U.K.B., K.S., G.I., D.D., G.L., T.R.F.), R Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, Maryland
| | - K Sudini
- From the Department of Diagnostic Radiology and Nuclear Medicine (U.K.B., K.S., G.I., D.D., G.L., T.R.F.), R Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, Maryland
| | - T R Fleiter
- From the Department of Diagnostic Radiology and Nuclear Medicine (U.K.B., K.S., G.I., D.D., G.L., T.R.F.), R Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, Maryland
| |
Collapse
|
57
|
Alford S, Patel D, Perakakis N, Mantzoros CS. Obesity as a risk factor for Alzheimer's disease: weighing the evidence. Obes Rev 2018; 19:269-280. [PMID: 29024348 DOI: 10.1111/obr.12629] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/03/2017] [Accepted: 09/04/2017] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the sixth leading cause of death in the USA today; therefore, it is imperative that public health initiatives and clinical strategies are developed to prevent and effectively treat AD. Despite the enormous impact that AD has on individuals, families, society, and the health care system, there are no biomarkers to clearly identify those at risk for AD, public health prevention strategies in place, or treatments to address the underlying pathology or stop the progression of AD. There is ample scientific as well as empirical evidence that obesity and its metabolic and vascular comorbidities are related to AD and likely in the causative pathway. Obesity prevention and treatment could prove to be an efficacious and safe approach to preventing AD, a serious and daunting epidemic disease. In this review, we present the current pathophysiological and clinical evidence linking obesity and obesity-related comorbidities (eg, insulin resistance, hyperglycaemia, and type 2 diabetes) with AD. Additionally, we discuss which population to target and when to consider treatment for AD. Finally, we summarize the current evidence regarding the efficacy of anti-obesity and anti-diabetic pharmacotherapeutic agents for the treatment of AD.
Collapse
Affiliation(s)
| | - D Patel
- MCPHS University, Boston, MA, USA.,VA Boston Healthcare System, Boston, MA, USA
| | - N Perakakis
- Mantzoros Lab, Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - C S Mantzoros
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
58
|
Lee JY, Choi HY, Park CS, Ju BG, Yune TY. Mithramycin A Improves Functional Recovery by Inhibiting BSCB Disruption and Hemorrhage after Spinal Cord Injury. J Neurotrauma 2017; 35:508-520. [PMID: 29048243 DOI: 10.1089/neu.2017.5235] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
After spinal cord injury (SCI), blood-spinal cord barrier (BSCB) disruption and progressive hemorrhage lead to secondary injury, subsequent apoptosis and/or necrosis of neurons and glia, causing permanent neurological deficits. Growing evidence indicates that mithramycin A (MA), an anti-cancer drug, has neuroprotective effects in ischemic brain injury and Huntington's disease (HD). However, the precise mechanism underlying its protective effects is largely unknown. Here, we examined the effect of MA on BSCB breakdown and hemorrhage as well as subsequent inflammation after SCI. After moderate spinal cord contusion injury at T9, MA (150 μg/kg) was immediately injected intraperitoneally (i.p.) and further injected once a day for 5 days. Our data show that MA attenuated BSCB disruption and hemorrhage, and inhibited the infiltration of neutrophils and macrophages after SCI. Consistent with these findings, the expression of inflammatory mediators was significantly alleviated by MA. MA also inhibited the expression and activation of matrix metalloprotease-9 (MMP-9) after injury, which is known to disrupt BSCB and the degradation of tight junction (TJ) proteins. In addition, the expression of sulfonylurea receptor 1 (SUR1) and transient receptor potential melastatin 4 (TRPM4), which are known to mediate hemorrhage at an early stage after SCI, was significantly blocked by MA treatment. Finally, MA inhibited apoptotic cell death and improved functional recovery after injury. Thus, our results demonstrated that MA improves functional recovery by attenuating BSCB disruption and hemorrhage through the downregulation of SUR1/TRPM4 and MMP-9 after SCI.
Collapse
Affiliation(s)
- Jee Y Lee
- 1 Age-Related and Brain Diseases Research Center, School of Medicine, Kyung Kyung Hee University , Seoul, Republic of Korea
| | - Hae Y Choi
- 1 Age-Related and Brain Diseases Research Center, School of Medicine, Kyung Kyung Hee University , Seoul, Republic of Korea
| | - Chan S Park
- 2 KHU-KIST Department of Converging Science and Technology, School of Medicine, Kyung Kyung Hee University , Seoul, Republic of Korea
| | - Bong G Ju
- 3 Department of Life Science, Sogang University , Seoul, Republic of Korea
| | - Tae Y Yune
- 1 Age-Related and Brain Diseases Research Center, School of Medicine, Kyung Kyung Hee University , Seoul, Republic of Korea
- 2 KHU-KIST Department of Converging Science and Technology, School of Medicine, Kyung Kyung Hee University , Seoul, Republic of Korea
- 4 Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Kyung Hee University , Seoul, Republic of Korea
| |
Collapse
|
59
|
Jha RM, Puccio AM, Okonkwo DO, Zusman BE, Park SY, Wallisch J, Empey PE, Shutter LA, Clark RSB, Kochanek PM, Conley YP. ABCC8 Single Nucleotide Polymorphisms are Associated with Cerebral Edema in Severe TBI. Neurocrit Care 2017; 26:213-224. [PMID: 27677908 DOI: 10.1007/s12028-016-0309-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Cerebral edema (CE) in traumatic brain injury (TBI) is the consequence of multiple underlying mechanisms and is associated with unfavorable outcomes. Genetic variability in these pathways likely explains some of the clinical heterogeneity observed in edema development. A role for sulfonylurea receptor-1 (Sur1) in CE is supported. However, there are no prior studies examining the effect of genetic variability in the Sur1 gene (ABCC8) on the development of CE. We hypothesize that ABCC8 single nucleotide polymorphisms (SNPs) are predictive of CE. METHODS DNA was extracted from 385 patients. SNPs in ABCC8 were genotyped using the Human Core Exome v1.2 (Illumina). CE measurements included acute CT edema, mean and peak intracranial pressure (ICP), and need for decompressive craniotomy. RESULTS Fourteen SNPs with minor allele frequency >0.2 were identified. Four SNPS rs2283261, rs3819521, rs2283258, and rs1799857 were associated with CE measures. In multiple regression models, homozygote-variant genotypes in rs2283261, rs3819521, and rs2283258 had increased odds of CT edema (OR 2.45, p = 0.007; OR 2.95, p = 0.025; OR 3.00, p = 0.013), had higher mean (β = 3.13, p = 0.000; β = 2.95, p = 0.005; β = 3.20, p = 0.008), and peak ICP (β = 8.00, p = 0.001; β = 7.64, p = 0.007; β = 6.89, p = 0.034). The homozygote wild-type genotype of rs1799857 had decreased odds of decompressive craniotomy (OR 0.47, p = 0.004). CONCLUSIONS This is the first report assessing the impact of ABCC8 genetic variability on CE development in TBI. Minor allele ABCC8 SNP genotypes had increased risk of CE, while major SNP alleles were protective-potentially suggesting an evolutionary advantage. These findings could guide risk stratification, treatment responders, and the development of novel targeted or gene-based therapies against CE in TBI and other neurological disorders.
Collapse
Affiliation(s)
- Ruchira M Jha
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA, 15261, USA. .,Department of Neurosurgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA. .,Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA. .,Safar Center for Resuscitation Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA. .,Clinical and Translational Science Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Ava M Puccio
- Department of Neurosurgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - David O Okonkwo
- Department of Neurosurgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Benjamin E Zusman
- Department of Neurosurgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Seo-Young Park
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jessica Wallisch
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA, 15261, USA.,Safar Center for Resuscitation Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Philip E Empey
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA.,Clinical and Translational Science Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lori A Shutter
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA, 15261, USA.,Department of Neurosurgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert S B Clark
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA, 15261, USA.,Safar Center for Resuscitation Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Anesthesiology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patrick M Kochanek
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA, 15261, USA.,Safar Center for Resuscitation Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Anesthesiology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Clinical and Translational Science Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yvette P Conley
- Clinical and Translational Science Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,School of Nursing, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
60
|
Leiva-Salcedo E, Riquelme D, Cerda O, Stutzin A. TRPM4 activation by chemically- and oxygen deprivation-induced ischemia and reperfusion triggers neuronal death. Channels (Austin) 2017; 11:624-635. [PMID: 28876976 DOI: 10.1080/19336950.2017.1375072] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cerebral ischemia-reperfusion injury triggers a deleterious process ending in neuronal death. This process has two components, a glutamate-dependent and a glutamate-independent mechanism. In the glutamate-independent mechanism, neurons undergo a slow depolarization eventually leading to neuronal death. However, little is known about the molecules that take part in this process. Here we show by using mice cortical neurons in culture and ischemia-reperfusion protocols that TRPM4 is fundamental for the glutamate-independent neuronal damage. Thus, by blocking excitotoxicity, we reveal a slow activating, glibenclamide- and 9-phenanthrol-sensitive current, which is activated within 5 min upon ischemia-reperfusion onset. TRPM4 shRNA-based silenced neurons show a reduced ischemia-reperfusion induced current and depolarization. Neurons were protected from neuronal death up to 3 hours after the ischemia-reperfusion challenge. The activation of TRPM4 during ischemia-reperfusion injury involves the increase in both, intracellular calcium and H2O2, which may act together to produce a sustained activation of the channel.
Collapse
Affiliation(s)
- Elías Leiva-Salcedo
- a Departamento de Biología , Facultad de Química y Biología, Universidad de Santiago de Chile , Santiago , Chile
| | - Denise Riquelme
- a Departamento de Biología , Facultad de Química y Biología, Universidad de Santiago de Chile , Santiago , Chile
| | - Oscar Cerda
- b Programa de Biología Celular y Molecular , Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile , Santiago , Chile.,c Millennium Nucleus of Ion Channels-Associated Diseases (MiNICAD) , Universidad de Chile , Santiago , Chile
| | - Andrés Stutzin
- d Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile , Santiago , Chile
| |
Collapse
|
61
|
Adams SM, Conley YP, Wagner AK, Jha RM, Clark RSB, Poloyac SM, Kochanek PM, Empey PE. The pharmacogenomics of severe traumatic brain injury. Pharmacogenomics 2017; 18:1413-1425. [PMID: 28975867 PMCID: PMC5694019 DOI: 10.2217/pgs-2017-0073] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 07/06/2017] [Indexed: 01/08/2023] Open
Abstract
Pharmacotherapy for traumatic brain injury (TBI) is focused on resuscitation, prevention of secondary injury, rehabilitation and recovery. Pharmacogenomics may play a role in TBI for predicting therapies for sedation, analgesia, seizure prevention, intracranial pressure-directed therapy and neurobehavioral/psychiatric symptoms. Research into genetic predictors of outcomes and susceptibility to complications may also help clinicians to tailor therapeutics for high-risk individuals. Additionally, the expanding use of genomics in the drug development pipeline has provided insight to novel investigational and repurposed medications that may be useful in the treatment of TBI and its complications. Genomics in the context of treatment and prognostication for patients with TBI is a promising area for clinical progress of pharmacogenomics.
Collapse
Affiliation(s)
- Solomon M Adams
- Department of Pharmaceutical Sciences, Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Clinical & Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yvette P Conley
- Health Promotion & Development, School of Nursing, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Amy K Wagner
- Department of Physical Medicine & Rehabilitation, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Ruchira M Jha
- Clinical & Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, USA
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Neurological Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Robert SB Clark
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, USA
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Division of Pediatric Critical Care Medicine, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224, USA
| | - Samuel M Poloyac
- Department of Pharmaceutical Sciences, Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Clinical & Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Patrick M Kochanek
- Clinical & Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, USA
- Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Philip E Empey
- Clinical & Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA 15224, USA
- Department of Pharmacy & Therapeutics, Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
62
|
Maegele M, Schöchl H, Menovsky T, Maréchal H, Marklund N, Buki A, Stanworth S. Coagulopathy and haemorrhagic progression in traumatic brain injury: advances in mechanisms, diagnosis, and management. Lancet Neurol 2017; 16:630-647. [PMID: 28721927 DOI: 10.1016/s1474-4422(17)30197-7] [Citation(s) in RCA: 204] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 05/08/2017] [Accepted: 05/30/2017] [Indexed: 01/28/2023]
Abstract
Normal haemostasis depends on an intricate balance between mechanisms of bleeding and mechanisms of thrombosis, and this balance can be altered after traumatic brain injury (TBI). Impaired haemostasis could exacerbate the primary insult with risk of initiation or aggravation of bleeding; anticoagulant use at the time of injury can also contribute to bleeding risk after TBI. Many patients with TBI have abnormalities on conventional coagulation tests at admission to the emergency department, and the presence of coagulopathy is associated with increased morbidity and mortality. Further blood testing often reveals a range of changes affecting platelet numbers and function, procoagulant or anticoagulant factors, fibrinolysis, and interactions between the coagulation system and the vascular endothelium, brain tissue, inflammatory mechanisms, and blood flow dynamics. However, the degree to which these coagulation abnormalities affect TBI outcomes and whether they are modifiable risk factors are not known. Although the main challenge for management is to address the risk of hypocoagulopathy with prolonged bleeding and progression of haemorrhagic lesions, the risk of hypercoagulopathy with an increased prothrombotic tendency also warrants consideration.
Collapse
Affiliation(s)
- Marc Maegele
- Department for Trauma and Orthopaedic Surgery, Cologne-Merheim Medical Center, University Witten/Herdecke, Cologne, Germany; Institute for Research in Operative Medicine, University Witten/Herdecke, Cologne, Germany.
| | - Herbert Schöchl
- Department for Anaesthesiology and Intensive Care Medicine, AUVA Trauma Academic Teaching Hospital, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Tomas Menovsky
- Department for Neurosurgery, Antwerp University Hospital, University of Antwerp, Edegem, Belgium
| | - Hugues Maréchal
- Department of Anaesthesiology and Intensive Care Medicine, CRH La Citadelle, Liège, Belgium
| | - Niklas Marklund
- Department of Clinical Sciences, Division of Neurosurgery, University Hospital of Southern Sweden, Lund University, Lund, Sweden
| | - Andras Buki
- Department of Neurosurgery, The MTA-PTE Clinical Neuroscience MR Research Group, Janos Szentagothai Research Center, Hungarian Brain Research Program, University of Pécs, Pécs, Hungary
| | - Simon Stanworth
- NHS Blood and Transplant/Oxford University Hospitals NHS Foundation Trust, University of Oxford, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
63
|
Stokum JA, Keledjian K, Hayman E, Karimy JK, Pampori A, Imran Z, Woo SK, Gerzanich V, Simard JM. Glibenclamide pretreatment protects against chronic memory dysfunction and glial activation in rat cranial blast traumatic brain injury. Behav Brain Res 2017; 333:43-53. [PMID: 28662892 DOI: 10.1016/j.bbr.2017.06.038] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/20/2017] [Accepted: 06/24/2017] [Indexed: 02/03/2023]
Abstract
Blast traumatic brain injury (bTBI) affects both military and civilian populations, and often results in chronic deficits in cognition and memory. Chronic glial activation after bTBI has been linked with cognitive decline. Pharmacological inhibition of sulfonylurea receptor 1 (SUR1) with glibenclamide was shown previously to reduce glial activation and improve cognition in contusive models of CNS trauma, but has not been examined in bTBI. We postulated that glibenclamide would reduce chronic glial activation and improve long-term memory function after bTBI. Using a rat direct cranial model of bTBI (dc-bTBI), we evaluated the efficacy of two glibenclamide treatment paradigms: glibenclamide prophylaxis (pre-treatment), and treatment with glibenclamide starting after dc-bTBI (post-treatment). Our results show that dc-bTBI caused hippocampal astrocyte and microglial/macrophage activation that was associated with hippocampal memory dysfunction (rapid place learning paradigm) at 28days, and that glibenclamide pre-treatment, but not post-treatment, effectively protected against glial activation and memory dysfunction. We also report that a brief transient time-window of blood-brain barrier (BBB) disruption occurs after dc-bTBI, and we speculate that glibenclamide, which is mostly protein bound and does not normally traverse the intact BBB, can undergo CNS delivery only during this brief transient opening of the BBB. Together, our findings indicate that prophylactic glibenclamide treatment may help to protect against chronic cognitive sequelae of bTBI in warfighters and other at-risk populations.
Collapse
Affiliation(s)
- Jesse A Stokum
- Departments of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD 21201, USA.
| | - Kaspar Keledjian
- Departments of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD 21201, USA
| | - Erik Hayman
- Departments of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD 21201, USA
| | - Jason K Karimy
- Departments of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD 21201, USA
| | - Adam Pampori
- Departments of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD 21201, USA
| | - Ziyan Imran
- Departments of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD 21201, USA
| | - Seung Kyoon Woo
- Departments of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD 21201, USA
| | - Volodymyr Gerzanich
- Departments of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD 21201, USA
| | - J Marc Simard
- Departments of Pathology, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD 21201, USA; Departments of Physiology, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD 21201, USA
| |
Collapse
|
64
|
Effects of Oral Glibenclamide on Brain Contusion Volume and Functional Outcome of Patients with Moderate and Severe Traumatic Brain Injuries: A Randomized Double-Blind Placebo-Controlled Clinical Trial. World Neurosurg 2017; 101:130-136. [DOI: 10.1016/j.wneu.2017.01.103] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 01/19/2017] [Accepted: 01/26/2017] [Indexed: 01/28/2023]
|
65
|
FVIIa prevents the progressive hemorrhaging of a brain contusion by protecting microvessels via formation of the TF–FVIIa–FXa complex. Neuroscience 2017; 348:114-125. [DOI: 10.1016/j.neuroscience.2017.02.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 01/20/2017] [Accepted: 02/12/2017] [Indexed: 11/20/2022]
|
66
|
Jha RM, Puccio AM, Chou SHY, Chang CCH, Wallisch JS, Molyneaux BJ, Zusman BE, Shutter LA, Poloyac SM, Janesko-Feldman KL, Okonkwo DO, Kochanek PM. Sulfonylurea Receptor-1: A Novel Biomarker for Cerebral Edema in Severe Traumatic Brain Injury. Crit Care Med 2017; 45:e255-e264. [PMID: 27845954 PMCID: PMC5550829 DOI: 10.1097/ccm.0000000000002079] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Cerebral edema is a key poor prognosticator in traumatic brain injury. There are no biomarkers identifying patients at-risk, or guiding mechanistically-precise therapies. Sulfonylurea receptor-1-transient receptor potential cation channel M4 is upregulated only after brain injury, causing edema in animal studies. We hypothesized that sulfonylurea receptor-1 is measurable in human cerebrospinal fluid after severe traumatic brain injury and is an informative biomarker of edema and outcome. DESIGN A total of 119 cerebrospinal fluid samples were collected from 28 severe traumatic brain injury patients. Samples were retrieved at 12, 24, 48, 72 hours and before external ventricular drain removal. Fifteen control samples were obtained from patients with normal pressure hydrocephalus. Sulfonylurea receptor- 1 was quantified by enzyme-linked immunosorbent assay. Outcomes included CT edema, intracranial pressure measurements, therapies targeting edema, and 3-month Glasgow Outcome Scale score. MAIN RESULTS Sulfonylurea receptor-1 was present in all severe traumatic brain injury patients (mean = 3.54 ± 3.39 ng/mL, peak = 7.13 ± 6.09 ng/mL) but undetectable in all controls (p < 0.001). Mean and peak sulfonylurea receptor-1 was higher in patients with CT edema (4.96 ± 1.13 ng/mL vs 2.10 ± 0.34 ng/mL; p = 0.023). There was a temporal delay between peak sulfonylurea receptor-1 and peak intracranial pressure in 91.7% of patients with intracranial hypertension. There was no association between mean/peak sulfonylurea receptor-1 and mean/peak intracranial pressure, proportion of intracranial pressure greater than 20 mm Hg, use of edema-directed therapies, decompressive craniotomy, or 3-month Glasgow Outcome Scale. However, decreasing sulfonylurea receptor-1 trajectories between 48 and 72 hours were significantly associated with improved cerebral edema and clinical outcome. Area under the multivariate model receiver operating characteristic curve was 0.881. CONCLUSIONS This is the first report quantifying human cerebrospinal fluid sulfonylurea receptor-1. Sulfonylurea receptor-1 was detected in severe traumatic brain injury, absent in controls, correlated with CT-edema and preceded peak intracranial pressure. Sulfonylurea receptor-1 trajectories between 48 and 72 hours were associated with outcome. Because a therapy inhibiting sulfonylurea receptor-1 is available, assessing cerebrospinal fluid sulfonylurea receptor-1 in larger studies is warranted to evaluate our exploratory findings regarding its diagnostic, and monitoring utility, as well as its potential to guide targeted therapies in traumatic brain injury and other diseases involving cerebral edema.
Collapse
Affiliation(s)
- Ruchira M Jha
- 1Department of Critical Care Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA. 2Department of Neurosurgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA. 3Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA. 4Safar Center for Resuscitation Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA. 5Clinical and Translational Science, School of Medicine, University of Pittsburgh, Pittsburgh, PA. 6Department of Biostatistics, School of Medicine, University of Pittsburgh, Pittsburgh, PA. 7Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA. 8Department of Anesthesiology, School of Medicine, University of Pittsburgh, Pittsburgh, PA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Tsarenko SV, Dzyadz'ko AM, Rybalko SS. [Glibenclamide as a promising agent for prevention and treatment of cerebral edema]. ZHURNAL VOPROSY NEIROKHIRURGII IMENI N. N. BURDENKO 2017; 81:88-93. [PMID: 28665392 DOI: 10.17116/neiro201781388-93] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The article presents a review of the literature on the use of a fundamentally new technique for prevention and treatment of cerebral edema. A drug glibenclamide, which is used to treat type 2 diabetes mellitus, is able to reduce cerebral edema and neuronal damage as evidenced by the results of preclinical trials in rodents and the first results of drug application in patients. The article describes the mechanism of glibenclamide action and discusses the potential for its application.
Collapse
Affiliation(s)
- S V Tsarenko
- Treatment and Rehabilitation Center, Moscow, Russia
| | - A M Dzyadz'ko
- Republican Scientific and Practical Center of Organ and Tissue Transplantation, Minsk, Republic of Belarus
| | - S S Rybalko
- Republican Scientific and Practical Center of Neurology and Neurosurgery, Minsk, Republic of Belarus
| |
Collapse
|
68
|
Xu Z, Liu Y, Yang D, Yuan F, Ding J, Wang L, Qu M, Yang G, Tian H. Glibenclamide–sulfonylurea receptor 1 antagonist alleviates LPS-induced BV2 cell activation through the p38/MAPK pathway. RSC Adv 2017. [DOI: 10.1039/c7ra03042h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We investigated the anti-neuroinflammatory activity and mechanism of glibenclamide, sulfonylurea receptor 1 (Sur1) antagonist, against LPS-induced microglial activationin vitro.
Collapse
Affiliation(s)
- Zhiming Xu
- Department of Neurosurgery
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital
- Shanghai
- China
| | - Yingliang Liu
- Department of Neurosurgery
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital
- Shanghai
- China
| | - Dianxu Yang
- Department of Neurosurgery
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital
- Shanghai
- China
| | - Fang Yuan
- Department of Neurosurgery
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital
- Shanghai
- China
| | - Jun Ding
- Department of Neurosurgery
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital
- Shanghai
- China
| | - Liping Wang
- Department of Neurology
- Ruijin Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai
- China
| | - Meijie Qu
- Department of Neurology
- Ruijin Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai
- China
| | - Guoyuan Yang
- Department of Neurology
- Ruijin Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai
- China
| | - Hengli Tian
- Department of Neurosurgery
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital
- Shanghai
- China
| |
Collapse
|
69
|
Winkler EA, Minter D, Yue JK, Manley GT. Cerebral Edema in Traumatic Brain Injury. Neurosurg Clin N Am 2016; 27:473-88. [DOI: 10.1016/j.nec.2016.05.008] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
70
|
Bell ED, Donato AJ, Monson KL. Cerebrovascular dysfunction following subfailure axial stretch. J Mech Behav Biomed Mater 2016; 65:627-633. [PMID: 27736719 DOI: 10.1016/j.jmbbm.2016.09.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 09/17/2016] [Accepted: 09/21/2016] [Indexed: 11/28/2022]
Abstract
Cerebral blood vessels are vital to maintaining the health of the brain. Traumatic brain injury (TBI) commonly results in autoregulatory dysfunction and associated failure of cerebral vessels to maintain homeostasis in the brain. While post-injury changes to brain biochemistry are known to contribute to this dysfunction, tissue deformation may also directly alter vascular smooth muscle cell (SMC) function. As a first step toward understanding stretch-induced dysfunction, this study investigates the effect of overstretch on the contractile behavior of SMCs in middle cerebral arteries (MCAs). We hypothesized that vessel function is altered above a threshold of stretch and strain rate. Twenty-four MCAs from Sprague Dawley rats were tested. Following development of basal SMC tone, vessels were subjected to increasing levels of isosmotic extracellular potassium (K+). Samples were then subjected to an axial overstretch of either 1.2*λIV or 1.3*λIV at strain rates of 0.2 or 20s-1. Following overstretch, SMC contractile behavior was measured again, both immediately and 60min after overstretch. Control vessels were subjected to the same protocol but without overstretch. SMC contractile behavior was characterized using both percent contraction (%C) relative to the fully dilated inner diameter and the K+ dose required to evoke the half maximal contractile response (EC50). Control vessels exhibited increased sensitivity to K+ in successive characterization tests, so all effects were quantified relative to the time-matched control response. Samples exhibited the typical biphasic response to extracellular K+, dilating and contracting in response to small and large K+ concentrations, respectively. As hypothesized, axial overstretch altered SMC contractile behavior, as seen in a decrease in %C for sub-maximal contractile K+ doses (p<0.05) and an increase in EC50 (p<0.01), but only for the test group stretched rapidly to 1.3*λIV. While the change in %C was only significantly different immediately after overstretch, the change to EC50 persisted for 60min. These results indicate that deformation can alter SMC contractile behavior and thus potentially play a role in cerebrovascular autoregulatory dysfunction independent of the pathological chemical environment in the brain post-TBI.
Collapse
Affiliation(s)
- E David Bell
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA; Laboratory of Head Injury and Vessel Biomechanics, Department of Mechanical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Anthony J Donato
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, USA
| | - Kenneth L Monson
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA; Laboratory of Head Injury and Vessel Biomechanics, Department of Mechanical Engineering, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
71
|
Abstract
PURPOSE OF REVIEW Traumatic brain injury (TBI) remains the leading cause of morbidity and mortality in the United States. Over the last decade, several advancements have been made in the field of TBI all aimed at improving outcomes. RECENT FINDINGS Advancements in the management of TBI have been made possible through improved understanding of basic pathophysiology associated with this condition. The aim of this review is to briefly highlight the underlying pathophysiology of TBI and the most recent advancements and novel strategies being used in its treatment. We also briefly discuss coagulopathy of TBI, clinical management of TBI and how it has evolved recently. SUMMARY The mortality associated with TBI continues to remain high and several novel strategies have emerged as potential candidates for the treatment of secondary brain injury. The clinical management of TBI and associated coagulopathy has evolved allowing for a more tailored approach toward its management.
Collapse
|
72
|
Gainey SJ, Kwakwa KA, Bray JK, Pillote MM, Tir VL, Towers AE, Freund GG. Short-Term High-Fat Diet (HFD) Induced Anxiety-Like Behaviors and Cognitive Impairment Are Improved with Treatment by Glyburide. Front Behav Neurosci 2016; 10:156. [PMID: 27563288 PMCID: PMC4980396 DOI: 10.3389/fnbeh.2016.00156] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 07/29/2016] [Indexed: 01/21/2023] Open
Abstract
Obesity-associated comorbidities such as cognitive impairment and anxiety are increasing public health burdens that have gained prevalence in children. To better understand the impact of childhood obesity on brain function, mice were fed with a high-fat diet (HFD) from weaning for 1, 3 or 6 weeks. When compared to low-fat diet (LFD)-fed mice (LFD-mice), HFD-fed mice (HFD-mice) had impaired novel object recognition (NOR) after 1 week. After 3 weeks, HFD-mice had impaired NOR and object location recognition (OLR). Additionally, these mice displayed anxiety-like behavior by measure of both the open-field and elevated zero maze (EZM) testing. At 6 weeks, HFD-mice were comparable to LFD-mice in NOR, open-field and EZM performance but they remained impaired during OLR testing. Glyburide, a second-generation sulfonylurea for the treatment of type 2 diabetes, was chosen as a countermeasure based on previous data exhibiting its potential as an anxiolytic. Interestingly, a single dose of glyburide corrected deficiencies in NOR and mitigated anxiety-like behaviors in mice fed with HFD-diet for 3-weeks. Taken together these results indicate that a HFD negatively impacts a subset of hippocampal-independent behaviors relatively rapidly, but such behaviors normalize with age. In contrast, impairment of hippocampal-sensitive memory takes longer to develop but persists. Since single-dose glyburide restores brain function in 3-week-old HFD-mice, drugs that block ATP-sensitive K(+) (KATP) channels may be of clinical relevance in the treatment of obesity-associated childhood cognitive issues and psychopathologies.
Collapse
Affiliation(s)
- Stephen J Gainey
- Department of Animal Sciences, University of IllinoisUrbana, IL, USA; Department of Pathology, Program in Integrative Immunology and Behavior, University of Illinois College of MedicineUrbana, IL, USA
| | - Kristin A Kwakwa
- Department of Pathology, Program in Integrative Immunology and Behavior, University of Illinois College of Medicine Urbana, IL, USA
| | - Julie K Bray
- Department of Pathology, Program in Integrative Immunology and Behavior, University of Illinois College of Medicine Urbana, IL, USA
| | - Melissa M Pillote
- Department of Pathology, Program in Integrative Immunology and Behavior, University of Illinois College of Medicine Urbana, IL, USA
| | - Vincent L Tir
- Department of Pathology, Program in Integrative Immunology and Behavior, University of Illinois College of Medicine Urbana, IL, USA
| | - Albert E Towers
- Department of Pathology, Program in Integrative Immunology and Behavior, University of Illinois College of MedicineUrbana, IL, USA; Department of Nutritional Sciences, University of IllinoisUrbana, IL, USA
| | - Gregory G Freund
- Department of Animal Sciences, University of IllinoisUrbana, IL, USA; Department of Pathology, Program in Integrative Immunology and Behavior, University of Illinois College of MedicineUrbana, IL, USA; Department of Nutritional Sciences, University of IllinoisUrbana, IL, USA
| |
Collapse
|
73
|
Huang K, Wang Z, Gu Y, Hu Y, Ji Z, Wang S, Lin Z, Li X, Xie Z, Pan S. Glibenclamide Is Comparable to Target Temperature Management in Improving Survival and Neurological Outcome After Asphyxial Cardiac Arrest in Rats. J Am Heart Assoc 2016; 5:JAHA.116.003465. [PMID: 27413041 PMCID: PMC5015382 DOI: 10.1161/jaha.116.003465] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background We previously have shown that glibenclamide (GBC), a sulfonylurea receptor 1–transient receptor potential M4 (SUR1‐TRPM4) channel inhibitor, improves survival and neurological outcome after asphyxial cardiac arrest and cardiopulmonary resuscitation (ACA/CPR). Here, we further compare the efficacy of GBC with target temperature management (TTM) and determine whether the efficacy of GBC is affected by TTM. Methods and Results Male Sprague‐Dawley rats (n=213) subjected to 10‐minute ACA/CPR were randomized to 4 groups after return of spontaneous circulation (ROSC): normothermia control (NT); GBC; TTM; and TTM+GBC. Survival, neurodeficit scores, histological injury, as well as the expressions of SUR1 and TRPM4 were evaluated. The 7‐day survival rate was 34.4% (11 of 32) in the NT group, 65% (13 of 20) in the GBC group, 50% (10 of 20) in the TTM group, and 70% (14 of 20) in the TTM+GBC group. Rats that received either GBC, TTM alone, or in combination showed less neurological deficit than NT control at 24, 48, and 72 hours and 7 days after ROSC. Moreover, TTM or GBC ameliorated neuronal degeneration and glial activation in the hippocampal CA1 region with similar efficacy, whereas the combination of them had a trend toward better effect. The subunits of SUR1‐TRPM4 heterodimers were both strongly upregulated after ACA/CPR and expressed in multiple types of brain cells, but partly suppressed by TTM. Conclusions GBC is comparable to TTM in improving survival and neurological outcome after ACA/CPR. When GBC is given along with TTM, less histological injury tended to be achieved.
Collapse
Affiliation(s)
- Kaibin Huang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ziyue Wang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yong Gu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yafang Hu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhong Ji
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shengnan Wang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhenzhou Lin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xing Li
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zuoshan Xie
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Suyue Pan
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
74
|
Xu ZM, Yuan F, Liu YL, Ding J, Tian HL. Glibenclamide Attenuates Blood-Brain Barrier Disruption in Adult Mice after Traumatic Brain Injury. J Neurotrauma 2016; 34:925-933. [PMID: 27297934 DOI: 10.1089/neu.2016.4491] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Glibenclamide is a hypoglycemic drug that is widely used for the treatment of diabetes mellitus type 2 (DM II), but it also plays a protective role following injury to the central nervous system (CNS). However, the precise mechanisms underlying its neuroprotective actions remain to be elucidated. Therefore, the present study evaluated the effects of glibenclamide on the blood-brain barrier (BBB) in a mouse model of traumatic brain injury (TBI). In the present study, 86 adult male C57BL/6 mice were exposed to a controlled cortical impact (CCI) injury and then received glibenclamide (10 μg) for 3 days. Tight junction (TJ) protein levels, BBB permeability, and tissue hemoglobin levels were evaluated following the CCI injury. Additionally, a biaxial stretch injury was applied to cell cultures of bEnd.3 cells using the Cell Injury Controller II system to explore the mechanisms by which glibenclamide inhibits apoptosis-signaling pathways. Compared with the control group, glibenclamide-treated mice exhibited decreases in brain water content (p < 0.05), tissue hemoglobin levels (p < 0.05), and Evans Blue extravasation (p < 0.01) after the CCI injury. Glibenclamide primarily attenuated apoptosis via the JNK/c-jun signaling pathway and resulted in an elevation of stretch injury-induced ZO-1 expression in bEnd.3 cells (p < 0.01).Glibenclamide downregulated the activity of the JNK/c-jun apoptosis-signaling pathway which, in turn, decreased apoptosis in endothelial cells (ECs). This may have prevented the disruption of the BBB in a mouse model of TBI.
Collapse
Affiliation(s)
- Zhi-Ming Xu
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, China
| | - Fang Yuan
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, China
| | - Ying-Liang Liu
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, China
| | - Jun Ding
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, China
| | - Heng-Li Tian
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, China
| |
Collapse
|
75
|
Liu J, Tian HL. Relationship between trauma-induced coagulopathy and progressive hemorrhagic injury in patients with traumatic brain injury. Chin J Traumatol 2016; 19:172-5. [PMID: 27321300 PMCID: PMC4908229 DOI: 10.1016/j.cjtee.2016.01.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Progressive hemorrhagic injury (PHI) can be divided into coagulopathy-related PHI and normal coagu- lation PHI. Coagulation disorders after traumatic brain injuries can be included in trauma-induced coagulopathy (TIC). Some studies showed that TIC is associated with PHI and increases the rates of disability and mortality. In this review, we discussed some mechanisms in TIC, which is of great importance in the development of PHI, including tissue factor (TF) hypothesis, protein C pathway and thrombocytopenia. The main mechanism in the relation of TIC to PHI is hypocoagulability. We also reviewed some coagulopathy parameters and proposed some possible risk factors, predictors and therapies.
Collapse
Affiliation(s)
| | - Heng-Li Tian
- Corresponding author. Tel.: +86 21 24058405; fax: +86 21 64369181.
| |
Collapse
|
76
|
Zafardoost P, Ghasemi AA, Salehpour F, Piroti C, Ziaeii E. Evaluation of the Effect of Glibenclamide in Patients With Diffuse Axonal Injury Due to Moderate to Severe Head Trauma. Trauma Mon 2016; 21:e25113. [PMID: 28184360 PMCID: PMC5292034 DOI: 10.5812/traumamon.25113] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 03/13/2015] [Accepted: 03/29/2015] [Indexed: 02/07/2023] Open
Abstract
Background Traumatic brain injury (TBI) is a major health problem worldwide. Secondary injuries after TBI, including diffuse axonal injury (DAI) often occur, and proper treatments are needed in this regard. It has been shown that glibenclamide could reduce secondary brain damage after experimental TBI and improve outcomes. Objectives We aim to evaluate the role of glibenclamide on the short-term outcome of patients with DAI due to moderate to severe TBI. Patients and Methods In this controlled randomized clinical trial, 40 patients with moderate to severe TBI were assigned to glibenclamide (n = 20) and control (n = 20) groups. Six hours after admission the intervention group received 1.25 mg glibenclamide every 12 hours. The Glasgow coma scale (GCS) was administered at admission, in the first 24 and 48 hours, at one week post-trauma and at discharge. The Glasgow outcome scale (GOS) was also administered at discharge. All results were evaluated and compared between groups. Results Patients treated with glibenclamide compared to the control group had a significantly better GCS score one week post-trauma (P = 0.003) and at discharge (P = 0.004), as well as a better GOS score at discharge (P = 0.001). The glibenclamide group also had a shorter length of hospital stay compared to the control group (P = 0.03). In the control group, two patients (10%) died during the first week post-trauma, but there was no mortality in the glibenclamide group (P = 0.48). Conclusions Treatment with glibenclamide in patients with DAI due to moderate to severe TBI significantly improves short-term outcomes.
Collapse
Affiliation(s)
- Peyman Zafardoost
- Neurosurgery Department, Urmia University of Medical Sciences, Urmia, IR Iran
- Corresponding author: Peyman Zafardoost, Neurosurgery Department, Urmia University of Medical Sciences, Urmia, IR Iran. Tel: +98-4433485324, E-mail:
| | - Amir Abbas Ghasemi
- Neurosurgery Department, Urmia University of Medical Sciences, Urmia, IR Iran
| | - Firooz Salehpour
- Neurosurgery Department, Urmia University of Medical Sciences, Urmia, IR Iran
| | - Chia Piroti
- Neurosurgery Department, Urmia University of Medical Sciences, Urmia, IR Iran
| | - Ehsan Ziaeii
- Neurosurgery Department, Urmia University of Medical Sciences, Urmia, IR Iran
| |
Collapse
|
77
|
Welling LC, Welling MS, Teixeira MJ, Figueiredo EG. Brain Contusions and SUR1 Receptors: Has a New Target Been Born? World Neurosurg 2016; 87:475-6. [DOI: 10.1016/j.wneu.2016.01.044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
78
|
Kenney K, Amyot F, Haber M, Pronger A, Bogoslovsky T, Moore C, Diaz-Arrastia R. Cerebral Vascular Injury in Traumatic Brain Injury. Exp Neurol 2016; 275 Pt 3:353-366. [DOI: 10.1016/j.expneurol.2015.05.019] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 05/19/2015] [Accepted: 05/26/2015] [Indexed: 12/14/2022]
|
79
|
Hanna A, Boggs DH, Kwok Y, Simard M, Regine WF, Mehta M. What predicts early volumetric edema increase following stereotactic radiosurgery for brain metastases? J Neurooncol 2015; 127:303-11. [DOI: 10.1007/s11060-015-2034-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 12/26/2015] [Indexed: 10/22/2022]
|
80
|
Ernstbrunner L, Korn G, Ernstbrunner E, Auffarth A, Tauber M, Resch H, Moroder P. S100B serum protein cannot predict secondary intracranial haemorrhage after mild head injury in patients with low-dose acetylsalicylic acid prophylaxis. Brain Inj 2015; 30:43-7. [PMID: 26580090 DOI: 10.3109/02699052.2015.1087593] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
INTRODUCTION The goal of this study was to investigate if S100B serum protein could predict secondary intracranial haemorrhagic events (SIHEs) after mild head injury (mHI) in patients taking low-dose acetylsalicylic acid (LDA), making routinely repeated head computed tomography (RRHCT) scans unnecessary. METHODS Three hundred and eight-two patients with mHI, older than 60 years and taking LDA prophylaxis were enrolled. Primary head CT and RRHCT scans within 3 and 48 hours to trauma were performed. Additionally, S100B serum protein levels were evaluated at admission and predictive power for SIHEs was analysed. RESULTS Fifty-nine per cent were female and the mean age of all included patients was 81.8 ± 8.9 years. In four patients SIHEs were diagnosed. Sensitivity and the negative predictive value of S100B serum protein (cut-off value 0.10 µg l(-1)) were 75.0% and 98.6%, respectively. Specificity was 19.0% and the positive predictive value 1.0% (306 false positive values). In patients without bleeding, the median S100B value was 0.18 (IQR = 0.12-0.34) and in the ones with SIHEs, the median was 0.11 (IQR = 0.10-1.16) (p > 0.05). The discriminatory power of S100B in the ROC analysis was 0.399 (95% CI = 0.079-0.720; p > 0.05). CONCLUSION S100B cannot be considered as an effective diagnostic tool in the prediction or exclusion of SIHE in older patients with mHIs taking LDA prophylaxis.
Collapse
Affiliation(s)
- Lukas Ernstbrunner
- a Department of Traumatology and Sports Injuries , Paracelsus Medical University , Salzburg , Austria
| | - Gundobert Korn
- a Department of Traumatology and Sports Injuries , Paracelsus Medical University , Salzburg , Austria
| | | | - Alexander Auffarth
- a Department of Traumatology and Sports Injuries , Paracelsus Medical University , Salzburg , Austria
| | - Mark Tauber
- a Department of Traumatology and Sports Injuries , Paracelsus Medical University , Salzburg , Austria .,c Department of Shoulder and Elbow Surgery , Atos Clinic , Munich , Germany
| | - Herbert Resch
- a Department of Traumatology and Sports Injuries , Paracelsus Medical University , Salzburg , Austria
| | - Philipp Moroder
- a Department of Traumatology and Sports Injuries , Paracelsus Medical University , Salzburg , Austria
| |
Collapse
|
81
|
Rosa M, da Rocha AJ, Maia ACM, Saade N, Veiga JCE, Romero JM. Contusion Contrast Extravasation Depicted on Multidetector Computed Tomography Angiography Predicts Growth and Mortality in Traumatic Brain Contusion. J Neurotrauma 2015. [PMID: 26214242 DOI: 10.1089/neu.2015.4062] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Traumatic brain injury (TBI) is the main cause of death in trauma victims and causes high rates of disability and neurological sequelae. Approximately 38-65% of traumatic brain contusions (TBC) demonstrate hemorrhagic expansion on serial computed tomography (CT) scans. Thus far, however, no single variable can accurately predict the hemorrhage expansion of a TBC. Our purpose was to evaluate contrast extravasation (CE) as a predictor of expansion, mortality, and poor outcome in TBC in a Brazilian cohort. After Institutional Review Board approval, we used multidetector CT angiography (MDCTA) to study 121 consecutive patients (106 men, 87.6%) with ages varying from 10 to 85 years. Informed consent was obtained from all subjects. The clinical and imaging findings were correlated with the findings on the initial MDCTA using either the Fisher exact test or Student t test and a multivariate logistic regression model. Of the persons who presented CE in TBC, 21.8% died (in-hospital mortality), whereas in the absence of this sign, the mortality rate was 7.6% (p = 0.014). In addition, expansion of the hemorrhagic component of the TBC was detected in 61.1% of the CE-positive patients, whereas expansion was only observed in 10% of the CE-negative patients (p < 0.001). Poor outcome was observed in 24.2% of the patients in the CE-negative group, but in the presence of CE, 72.7% evolved with poor outcome (p < 0.001). The CE was a strong independent predictor of expansion, poor outcome, and increased risk of in-hospital mortality in our series of patients with TBC.
Collapse
Affiliation(s)
- Marcos Rosa
- 1 Section of Neuroradiology , Santa Casa de Misericórdia de São Paulo, São Paulo SP, Brazil .,2 Section of Radiology, Federal University of Espírito Santo , Vitória ES, Brazil
| | - Antônio José da Rocha
- 1 Section of Neuroradiology , Santa Casa de Misericórdia de São Paulo, São Paulo SP, Brazil
| | | | - Nelson Saade
- 3 Division of Neurosurgery , Santa Casa de Misericórdia de São Paulo, São Paulo SP, Brazil
| | | | - Javier M Romero
- 4 Division of Neuroradiology, Massachusetts General Hospital , Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
82
|
Martínez-Valverde T, Vidal-Jorge M, Martínez-Saez E, Castro L, Arikan F, Cordero E, Rădoi A, Poca MA, Simard JM, Sahuquillo J. Sulfonylurea Receptor 1 in Humans with Post-Traumatic Brain Contusions. J Neurotrauma 2015; 32:1478-87. [PMID: 26398596 DOI: 10.1089/neu.2014.3706] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Post-traumatic brain contusions (PTBCs) are traditionally considered primary injuries and can increase in size, generate perilesional edema, cause mass effect, induce neurological deterioration, and cause death. Most patients experience a progressive increase in pericontusional edema, and nearly half, an increase in the hemorrhagic component itself. The underlying molecular pathophysiology of contusion-induced brain edema and hemorrhagic progression remains poorly understood. The aim of this study was to investigate sulfonylurea 1/transient receptor potential melastatin 4 (SUR1-TRPM4) ion channel SUR1 expression in various cell types (neurons, astrocytes, endothelial cells, microglia, macrophages, and neutrophils) of human brain contusions and whether SUR1 up-regulation was related to time postinjury. Double immunolabeling of SUR1 and cell-type- specific proteins was performed in 26 specimens from traumatic brain injury patients whose lesions were surgically evacuated. Three samples from limited brain resections performed for accessing extra-axial skull-base tumors or intraventricular lesions were controls. We found SUR1 was significantly overexpresed in all cell types and was especially prominent in neurons and endothelial cells (ECs). The temporal pattern depended on cell type: 1) In neurons, SUR1 increased within 48 h of injury and stabilized thereafter; 2) in ECs, there was no trend; 3) in glial cells and microglia/macrophages, a moderate increase was observed over time; and 4) in neutrophils, it decreased with time. Our results suggest that up-regulation of SUR1 in humans point to this channel as one of the important molecular players in the pathophysiology of PTBCs. Our findings reveal opportunities to act therapeutically on the mechanisms of growth of traumatic contusions and therefore reduce the number of patients with neurological deterioration and poor neurological outcomes.
Collapse
Affiliation(s)
- Tamara Martínez-Valverde
- 1 Neurotraumatology and Neurosurgery Research Unit (UNINN), Universitat Autònoma de Barcelona , Barcelona, Spain
| | - Marian Vidal-Jorge
- 1 Neurotraumatology and Neurosurgery Research Unit (UNINN), Universitat Autònoma de Barcelona , Barcelona, Spain
| | - Elena Martínez-Saez
- 2 Department of Pathology, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona , Barcelona, Spain
| | - Lidia Castro
- 1 Neurotraumatology and Neurosurgery Research Unit (UNINN), Universitat Autònoma de Barcelona , Barcelona, Spain
| | - Fuat Arikan
- 1 Neurotraumatology and Neurosurgery Research Unit (UNINN), Universitat Autònoma de Barcelona , Barcelona, Spain .,3 Department of Neurosurgery, Vall d'Hebron University Hospital , Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Esteban Cordero
- 3 Department of Neurosurgery, Vall d'Hebron University Hospital , Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Andreea Rădoi
- 1 Neurotraumatology and Neurosurgery Research Unit (UNINN), Universitat Autònoma de Barcelona , Barcelona, Spain
| | - Maria-Antonia Poca
- 1 Neurotraumatology and Neurosurgery Research Unit (UNINN), Universitat Autònoma de Barcelona , Barcelona, Spain .,3 Department of Neurosurgery, Vall d'Hebron University Hospital , Universitat Autònoma de Barcelona, Barcelona, Spain
| | - J Marc Simard
- 4 Departments of Neurosurgery, Physiology and Pathology, University of Maryland School of Medicine , Baltimore, Maryland
| | - Juan Sahuquillo
- 1 Neurotraumatology and Neurosurgery Research Unit (UNINN), Universitat Autònoma de Barcelona , Barcelona, Spain .,3 Department of Neurosurgery, Vall d'Hebron University Hospital , Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
83
|
|
84
|
Pathogenesis of brain edema and investigation into anti-edema drugs. Int J Mol Sci 2015; 16:9949-75. [PMID: 25941935 PMCID: PMC4463627 DOI: 10.3390/ijms16059949] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 04/15/2015] [Accepted: 04/27/2015] [Indexed: 12/18/2022] Open
Abstract
Brain edema is a potentially fatal pathological state that occurs after brain injuries such as stroke and head trauma. In the edematous brain, excess accumulation of extracellular fluid results in elevation of intracranial pressure, leading to impaired nerve function. Despite the seriousness of brain edema, only symptomatic treatments to remove edema fluid are currently available. Thus, the development of novel anti-edema drugs is required. The pathogenesis of brain edema is classified as vasogenic or cytotoxic edema. Vasogenic edema is defined as extracellular accumulation of fluid resulting from disruption of the blood-brain barrier (BBB) and extravasations of serum proteins, while cytotoxic edema is characterized by cell swelling caused by intracellular accumulation of fluid. Various experimental animal models are often used to investigate mechanisms underlying brain edema. Many soluble factors and functional molecules have been confirmed to induce BBB disruption or cell swelling and drugs targeted to these factors are expected to have anti-edema effects. In this review, we discuss the mechanisms and involvement of factors that induce brain edema formation, and the possibility of anti-edema drugs targeting them.
Collapse
|
85
|
Abstract
Traumatic brain injury (TBI) is a major cause of mortality and morbidity worldwide. Despite extensive preclinical research supporting the effectiveness of neuroprotective therapies for brain trauma, there have been no successful randomized controlled clinical trials to date. TBI results in delayed secondary tissue injury due to neurochemical, metabolic and cellular changes; modulating such effects has provided the basis for neuroprotective interventions. To establish more effective neuroprotective treatments for TBI it is essential to better understand the complex cellular and molecular events that contribute to secondary injury. Here we critically review relevant research related to causes and modulation of delayed tissue damage, with particular emphasis on cell death mechanisms and post-traumatic neuroinflammation. We discuss the concept of utilizing multipotential drugs that target multiple secondary injury pathways, rather than more specific "laser"-targeted strategies that have uniformly failed in clinical trials. Moreover, we assess data supporting use of neuroprotective drugs that are currently being evaluated in human clinical trials for TBI, as well as promising emerging experimental multipotential drug treatment strategies. Finally, we describe key challenges and provide suggestions to improve the likelihood of successful clinical translation.
Collapse
Affiliation(s)
- David J Loane
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bogdan A Stoica
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alan I Faden
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
86
|
McGinn MJ, Povlishock JT. Cellular and molecular mechanisms of injury and spontaneous recovery. HANDBOOK OF CLINICAL NEUROLOGY 2015; 127:67-87. [PMID: 25702210 DOI: 10.1016/b978-0-444-52892-6.00005-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Until recently, most have assumed that traumatic brain injury (TBI) was singularly associated with the overt destruction of brain tissue resulting in subsequent morbidity or death. More recently, experimental and clinical studies have shown that the pathobiology of TBI is more complex, involving a host of cellular and subcellular changes that impact on neuronal function and viability while also affecting vascular reactivity and the activation of multiple biological response pathways. Here we review the brain's response to injury, examining both focal and diffuse changes and their implications for post-traumatic brain dysfunction and recovery. TBI-induced neuronal dysfunction and death as well as the diffuse involvement of multiple fiber projections are discussed together with considerations of how local axonal membrane changes or channelopathy translate into local ionic dysregulation and axonal disconnection. Concomitant changes in the cerebral microcirculation are also discussed and their relationship with the parallel changes in the brain's metabolism is considered. These cellular and subcellular events occurring within neurons and their blood supply are correlated with multiple biological response modifiers evoked by generalized post-traumatic inflammation and the parallel activation of oxidative stress processes. The chapter closes with considerations of recovery following focal or diffuse injury. Evidence for dynamic brain reorganization/repair is presented, with considerations of traumatically induced circuit disruption and their progression to either adaptive or in some cases, maladaptive reorganization.
Collapse
Affiliation(s)
- Melissa J McGinn
- Department of Anatomy and Neurobiology, Medical College of Virginia Campus of Virginia Commonwealth University, Richmond, VA, USA
| | - John T Povlishock
- Department of Anatomy and Neurobiology, Medical College of Virginia Campus of Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
87
|
Diaz-Arrastia R, Kochanek PM, Bergold P, Kenney K, Marx CE, Grimes CJB, Loh LTCY, Adam LTCGE, Oskvig D, Curley KC, Salzer W. Pharmacotherapy of traumatic brain injury: state of the science and the road forward: report of the Department of Defense Neurotrauma Pharmacology Workgroup. J Neurotrauma 2014; 31:135-58. [PMID: 23968241 DOI: 10.1089/neu.2013.3019] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Despite substantial investments by government, philanthropic, and commercial sources over the past several decades, traumatic brain injury (TBI) remains an unmet medical need and a major source of disability and mortality in both developed and developing societies. The U.S. Department of Defense neurotrauma research portfolio contains more than 500 research projects funded at more than $700 million and is aimed at developing interventions that mitigate the effects of trauma to the nervous system and lead to improved quality of life outcomes. A key area of this portfolio focuses on the need for effective pharmacological approaches for treating patients with TBI and its associated symptoms. The Neurotrauma Pharmacology Workgroup was established by the U.S. Army Medical Research and Materiel Command (USAMRMC) with the overarching goal of providing a strategic research plan for developing pharmacological treatments that improve clinical outcomes after TBI. To inform this plan, the Workgroup (a) assessed the current state of the science and ongoing research and (b) identified research gaps to inform future development of research priorities for the neurotrauma research portfolio. The Workgroup identified the six most critical research priority areas in the field of pharmacological treatment for persons with TBI. The priority areas represent parallel efforts needed to advance clinical care; each requires independent effort and sufficient investment. These priority areas will help the USAMRMC and other funding agencies strategically guide their research portfolios to ensure the development of effective pharmacological approaches for treating patients with TBI.
Collapse
Affiliation(s)
- Ramon Diaz-Arrastia
- 1 Department of Neurology, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Zhang B, Gensel J. Is neuroinflammation in the injured spinal cord different than in the brain? Examining intrinsic differences between the brain and spinal cord. Exp Neurol 2014; 258:112-20. [DOI: 10.1016/j.expneurol.2014.04.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Revised: 03/28/2014] [Accepted: 04/08/2014] [Indexed: 12/17/2022]
|
89
|
Zweckberger K, Hackenberg K, Jung C, Hertle D, Kiening K, Unterberg A, Sakowitz O. Glibenclamide reduces secondary brain damage after experimental traumatic brain injury. Neuroscience 2014; 272:199-206. [DOI: 10.1016/j.neuroscience.2014.04.040] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 04/15/2014] [Accepted: 04/17/2014] [Indexed: 12/21/2022]
|
90
|
Nakadate H, Inuzuka K, Akanuma S, Kakuta A, Aomura S. Effect of amplitude and duration of impulsive pressure on endothelial permeability in in vitro fluid percussion trauma. Biomed Eng Online 2014; 13:44. [PMID: 24739360 PMCID: PMC4008480 DOI: 10.1186/1475-925x-13-44] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 04/04/2014] [Indexed: 01/07/2023] Open
Abstract
Background Intracranial pressure changes during head impact cause brain injuries such as vasogenic edema and cerebral contusion. However, the influence of impulsive pressure on endothelial function has not yet been fully studied in vitro. In this study, we developed a pressure loading device that produced positive and negative pressures by modifying an in vitro fluid percussion model and examined the effects of the amplitude and duration of the pressures on endothelial permeability. Methods Human umbilical vein endothelial cells were subjected to three types of positive pressure (average amplitude/average duration of 352 kPa/23 ms, 73 kPa/27 ms, and 70 kPa/44 ms) and three types of negative pressure (−72 kPa/41 ms, −67 kPa/104 ms, and −91 kPa/108 ms), and the transendothelial electrical resistance (TEER) was measured between 15 min and 24 h after pressure loading for quantifying the formation of an integral monolayer of endothelial cells. After loading, vascular endothelial- (VE-) cadherin, an endothelium-specific cell-cell adhesion molecule involved in endothelial barrier function, was stained and observed using fluorescence microscopy. Results The pressure loading device could produce positive pressure pulses with amplitudes of 53–1348 kPa and durations of 9–29.1 ms and negative pressure pulses with amplitudes of −52–−93 kPa and durations of 42.9–179.5 ms. The impulsive pressure reduced the TEER associated with the change in VE-cadherin localization. Additionally, TEER decreased considerably at 15 min and 6 h post-loading, with these changes being significant in positive pressure with larger amplitude and shorter duration and in all types of negative pressures compared to pre-loading. Conclusions The changes in intracranial pressure during head impact impair endothelial barrier function by the disruption of the integrity of endothelial cell-cell junctions, and the degree of increase in endothelial permeability depends on the amplitude, duration, and direction (compressive and tensile) of the impulsive pressure.
Collapse
Affiliation(s)
- Hiromichi Nakadate
- Graduate School of System Design, Tokyo Metropolitan University, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
91
|
Boggs DH, Simard JM, Steven A, Mehta MP. Potential of glyburide to reduce intracerebral edema in brain metastases. Expert Rev Neurother 2014; 14:379-88. [DOI: 10.1586/14737175.2014.890891] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
92
|
Iaccarino C, Schiavi P, Picetti E, Goldoni M, Cerasti D, Caspani M, Servadei F. Patients with brain contusions: predictors of outcome and relationship between radiological and clinical evolution. J Neurosurg 2014; 120:908-18. [PMID: 24506250 DOI: 10.3171/2013.12.jns131090] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Traumatic parenchymal mass lesions are common sequelae of traumatic brain injuries (TBIs). They occur in up to 8.2% of all TBI cases and 13%-35% of severe TBI cases, and they account for up to 20% of surgical intracranial lesions. Controversy exists concerning the association between radiological and clinical evolution of brain contusions. The aim of this study was to identify predictors of unfavorable outcome, analyze the evolution of brain contusions, and evaluate specific indications for surgery. METHODS In a retrospective, multicenter study, patients with brain contusions were identified in separate patient cohorts from 11 hospitals over a 4-year period (2008-2011). Data on clinical parameters and course of the contusion were collected. Radiological parameters were registered by using CT images taken at the time of hospital admission and at subsequent follow-up times. Patients who underwent surgical procedures were identified. Outcomes were evaluated 6 months after trauma by using the Glasgow Outcome Scale-Extended. RESULTS Multivariate analysis revealed the following reliable predictors of unfavorable outcome: 1) increased patient age, 2) lower Glasgow Coma Scale score at first evaluation, 3) clinical deterioration in the first hours after trauma, and 4) onset or increase of midline shift on follow-up CT images. Further multivariate analysis identified the following as statistically significant predictors of clinical deterioration during the first hours after trauma: 1) onset of or increase in midline shift on follow-up CT images (p < 0.001) and 2) increased effacement of basal cisterns on follow-up CT images (p < 0.001). CONCLUSIONS In TBI patients with cerebral contusion, the onset of clinical deterioration is predictably associated with the onset or increase of midline shift and worsened status of basal cisterns but not with hematoma or edema volume increase. A combination of clinical deterioration and increased midline shift/basal cistern compression is the most reasonable indicator for surgery.
Collapse
Affiliation(s)
- Corrado Iaccarino
- Arcispedale Santa Maria Nuova-Istituto di Ricovero e Cura a Carattere Scientifico, Reggio Emilia; and Neurosurgery-Neurotraumatology Unit
| | | | | | | | | | | | | |
Collapse
|
93
|
Kabadi SV, Faden AI. Neuroprotective strategies for traumatic brain injury: improving clinical translation. Int J Mol Sci 2014; 15:1216-36. [PMID: 24445258 PMCID: PMC3907865 DOI: 10.3390/ijms15011216] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 01/07/2014] [Accepted: 01/13/2014] [Indexed: 01/15/2023] Open
Abstract
Traumatic brain injury (TBI) induces secondary biochemical changes that contribute to delayed neuroinflammation, neuronal cell death, and neurological dysfunction. Attenuating such secondary injury has provided the conceptual basis for neuroprotective treatments. Despite strong experimental data, more than 30 clinical trials of neuroprotection in TBI patients have failed. In part, these failures likely reflect methodological differences between the clinical and animal studies, as well as inadequate pre-clinical evaluation and/or trial design problems. However, recent changes in experimental approach and advances in clinical trial methodology have raised the potential for successful clinical translation. Here we critically analyze the current limitations and translational opportunities for developing successful neuroprotective therapies for TBI.
Collapse
Affiliation(s)
- Shruti V Kabadi
- Department of Anesthesiology, Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Alan I Faden
- Department of Anesthesiology, Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
94
|
Inhibition of SUR1 decreases the vascular permeability of cerebral metastases. Neoplasia 2013; 15:535-43. [PMID: 23633925 DOI: 10.1593/neo.13164] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 03/04/2013] [Accepted: 03/11/2013] [Indexed: 01/13/2023] Open
Abstract
Inhibition of sulfonylurea receptor 1 (SUR1) by glyburide has been shown to decrease edema after subarachnoid hemorrhage. We investigated if inhibiting SUR1 reduces cerebral edema due to metastases, the most common brain tumor, and explored the putative association of SUR1 and the endothelial tight junction protein, zona occludens-1 (ZO-1). Nude rats were intracerebrally implanted with small cell lung carcinoma (SCLC) LX1 or A2058 melanoma cells (n = 36). Rats were administered vehicle, glyburide (4.8 µg twice, orally), or dexamethasone (0.35 mg, intravenous). Blood-tumor barrier (BTB) permeability (K (trans)) was evaluated before and after treatment using dynamic contrast-enhanced magnetic resonance imaging. SUR1 and ZO-1 expression was evaluated using immunofluorescence and Western blots. In both models, SUR1 expression was significantly increased (P < .05) in tumors. In animals with SCLC, control mean K (trans) (percent change ± standard error) was 101.8 ± 36.6%, and both glyburide (-21.4 ± 14.2%, P < .01) and dexamethasone (-14.2 ± 13.1%, P < .01) decreased BTB permeability. In animals with melanoma, compared to controls (117.1 ± 43.4%), glyburide lowered BTB permeability increase (3.2 ± 15.4%, P < .05), while dexamethasone modestly lowered BTB permeability increase (63.1 ± 22.1%, P > .05). Both glyburide (P < .001) and dexamethasone (P < .01) decreased ZO-1 gap formation. By decreasing ZO-1 gaps, glyburide was at least as effective as dexamethasone at halting increased BTB permeability caused by SCLC and melanoma. Glyburide is a safe, inexpensive, and efficacious alternative to dexamethasone for the treatment of cerebral metastasis-related vasogenic edema.
Collapse
|
95
|
Kurland DB, Tosun C, Pampori A, Karimy JK, Caffes NM, Gerzanich V, Simard JM. Glibenclamide for the treatment of acute CNS injury. Pharmaceuticals (Basel) 2013; 6:1287-303. [PMID: 24275850 PMCID: PMC3817601 DOI: 10.3390/ph6101287] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 09/17/2013] [Accepted: 09/23/2013] [Indexed: 12/22/2022] Open
Abstract
First introduced into clinical practice in 1969, glibenclamide (US adopted name, glyburide) is known best for its use in the treatment of diabetes mellitus type 2, where it is used to promote the release of insulin by blocking pancreatic KATP [sulfonylurea receptor 1 (Sur1)-Kir6.2] channels. During the last decade, glibenclamide has received renewed attention due to its pleiotropic protective effects in acute CNS injury. Acting via inhibition of the recently characterized Sur1-Trpm4 channel (formerly, the Sur1-regulated NCCa-ATP channel) and, in some cases, via brain KATP channels, glibenclamide has been shown to be beneficial in several clinically relevant rodent models of ischemic and hemorrhagic stroke, traumatic brain injury, spinal cord injury, neonatal encephalopathy of prematurity, and metastatic brain tumor. Glibenclamide acts on microvessels to reduce edema formation and secondary hemorrhage, it inhibits necrotic cell death, it exerts potent anti-inflammatory effects and it promotes neurogenesis—all via inhibition of Sur1. Two clinical trials, one in TBI and one in stroke, currently are underway. These recent findings, which implicate Sur1 in a number of acute pathological conditions involving the CNS, present new opportunities to use glibenclamide, a well-known, safe pharmaceutical agent, for medical conditions that heretofore had few or no treatment options.
Collapse
Affiliation(s)
- David B. Kurland
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (D.B.K.); (C.T.); (A.P.); (J.K.K.); (N.M.C.); (V.G.)
| | - Cigdem Tosun
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (D.B.K.); (C.T.); (A.P.); (J.K.K.); (N.M.C.); (V.G.)
| | - Adam Pampori
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (D.B.K.); (C.T.); (A.P.); (J.K.K.); (N.M.C.); (V.G.)
| | - Jason K. Karimy
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (D.B.K.); (C.T.); (A.P.); (J.K.K.); (N.M.C.); (V.G.)
| | - Nicholas M. Caffes
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (D.B.K.); (C.T.); (A.P.); (J.K.K.); (N.M.C.); (V.G.)
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (D.B.K.); (C.T.); (A.P.); (J.K.K.); (N.M.C.); (V.G.)
| | - J. Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA; E-Mails: (D.B.K.); (C.T.); (A.P.); (J.K.K.); (N.M.C.); (V.G.)
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-410-328-0850; Fax: +1-410-328-0124
| |
Collapse
|
96
|
Ding J, Yuan F, Guo JY, Chen H, Tian HL. Influence of glibenclamide on outcome in patients with type 2 diabetes and traumatic brain injury. Clin Neurol Neurosurg 2013; 115:2166-9. [PMID: 23998719 DOI: 10.1016/j.clineuro.2013.08.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Revised: 08/06/2013] [Accepted: 08/11/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND AND PURPOSE The influence of sulfonylurea receptor 1 (SUR1) and its inhibitor glibenclamide on progressive secondary hemorrhage (PSH), progressive hemorrhagic necrosis (PHN), and brain edema has been studied in rat models of traumatic brain injury (TBI) and ischemia. These studies indicate that blocking SUR1 may exert protective effects in terms of outcome. METHODS We discuss the effects of glibenclamide on outcome in patients with type 2 diabetes mellitus and TBI. We collected demographic, clinical, and imaging data from the clinical records of TBI patients with type 2 diabetes who were admitted to the neurosurgery department at Shanghai 6th People's Hospital between 2001 and 2012. Data from patients who met the inclusion criteria were analyzed. Patients were divided into glibenclamide group and insulin group. RESULTS Of 70 patients fit criteria for inclusion, no significant difference was observed except for age and fasting plasma glucose between the two groups. Outcome indicators, including GCS discharge, GOS discharge, length of study in hospital (LOS-H), and the presence of PSH showed no significant difference too (p>0.05), except for length of stay in neuro-intensive care unit (LOS-NICU) (p<0.05). Age, hours between the initial CT scan and the injury (HCT1) and GCS at admission were observed as factors associated with PSH after logistic regression. CONCLUSIONS In general, the use of glibenclamide to control plasma glucose after TBI had no significant effect on patient outcome at discharge but it could reduce the LOS-NICU (p<0.05). Glibenclamide also had no apparent effect on the presence of PSH in TBI patients with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Jun Ding
- Department of Neurosurgery, Shanghai Sixth People Hospital, Shanghai Jiaotong University, Shanghai 200233, China
| | | | | | | | | |
Collapse
|
97
|
Turtzo LC, Budde MD, Gold EM, Lewis BK, Janes L, Yarnell A, Grunberg NE, Watson W, Frank JA. The evolution of traumatic brain injury in a rat focal contusion model. NMR IN BIOMEDICINE 2013; 26:468-479. [PMID: 23225324 PMCID: PMC3596464 DOI: 10.1002/nbm.2886] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 08/28/2012] [Accepted: 10/05/2012] [Indexed: 06/01/2023]
Abstract
Serial MRI facilitates the in vivo analysis of the intra- and intersubject evolution of traumatic brain injury lesions. Despite the availability of MRI, the natural history of experimental focal contusion lesions in the controlled cortical impact (CCI) rat model has not been well described. We performed CCI on rats and MRI during the acute to chronic stages of cerebral injury to investigate the time course of changes in the brain. Female Wistar rats underwent CCI of their left motor cortex with a flat impact tip driven by an electromagnetic piston. In vivo MRI was performed at 7 T serially over 6 weeks post-CCI. The appearances of CCI-induced lesions and lesion-associated cortical volumes were variable on MRI, with the percentage change in cortical volume of the CCI ipsilateral side relative to the contralateral side ranging from 18% within 2 h of injury on day 0 to a peak of 35% on day 1, and a trough of -28% by week 5/6, with an average standard deviation of ± 14% at any given time point. In contrast, the percentage change in cortical volume of the ipsilateral side relative to the contralateral side in control rats was not significant (1 ± 2%). Hemorrhagic conversion within and surrounding the CCI lesion occurred between days 2 and 9 in 45% of rats, with no hemorrhage noted on the initial scan. Furthermore, hemorrhage and hemosiderin within the lesion were positive for Prussian blue and highly autofluorescent on histological examination. Although some variation in injuries may be technique related, the divergence of similar lesions between initial and final scans demonstrates the inherent biological variability of the CCI rat model.
Collapse
Affiliation(s)
- L. Christine Turtzo
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
| | - Matthew D. Budde
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
| | - Eric M. Gold
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
| | - Bobbi K. Lewis
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
| | - Lindsay Janes
- Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
| | - Angela Yarnell
- Department of Medical and Clinical Psychology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Neil E. Grunberg
- Department of Medical and Clinical Psychology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - William Watson
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Joseph A. Frank
- Frank Laboratory, National Institutes of Health, Bethesda, MD, USA
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
98
|
Rockswold SB, Rockswold GL, Zaun DA, Liu J. A prospective, randomized Phase II clinical trial to evaluate the effect of combined hyperbaric and normobaric hyperoxia on cerebral metabolism, intracranial pressure, oxygen toxicity, and clinical outcome in severe traumatic brain injury. J Neurosurg 2013; 118:1317-28. [PMID: 23510092 DOI: 10.3171/2013.2.jns121468] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECT Preclinical and clinical investigations indicate that the positive effect of hyperbaric oxygen (HBO2) for severe traumatic brain injury (TBI) occurs after rather than during treatment. The brain appears better able to use baseline O2 levels following HBO2 treatments. In this study, the authors evaluate the combination of HBO2 and normobaric hyperoxia (NBH) as a single treatment. METHODS Forty-two patients who sustained severe TBI (mean Glasgow Coma Scale [GCS] score 5.7) were prospectively randomized within 24 hours of injury to either: 1) combined HBO2/NBH (60 minutes of HBO2 at 1.5 atmospheres absolute [ATA] followed by NBH, 3 hours of 100% fraction of inspired oxygen [FiO2] at 1.0 ATA) or 2) control, standard care. Treatments occurred once every 24 hours for 3 consecutive days. Intracranial pressure, surrogate markers for cerebral metabolism, and O2 toxicity were monitored. Clinical outcome was assessed at 6 months using the sliding dichotomized Glasgow Outcome Scale (GOS) score. Mixed-effects linear modeling was used to statistically test differences between the treatment and control groups. Functional outcome and mortality rates were compared using chi-square tests. RESULTS There were no significant differences in demographic characteristics between the 2 groups. In comparison with values in the control group, brain tissue partial pressure of O2 (PO2) levels were significantly increased during and following combined HBO2/NBH treatments in both the noninjured and pericontusional brain (p < 0.0001). Microdialysate lactate/pyruvate ratios were significantly decreased in the noninjured brain in the combined HBO2/NBH group as compared with controls (p < 0.0078). The combined HBO2/NBH group's intracranial pressure values were significantly lower than those of the control group during treatment, and the improvement continued until the next treatment session (p < 0.0006). The combined HBO2/NBH group's levels of microdialysate glycerol were significantly lower than those of the control group in both noninjured and pericontusional brain (p < 0.001). The combined HBO2/NBH group's level of CSF F2-isoprostane was decreased at 6 hours after treatment as compared with that of controls, but the difference did not quite reach statistical significance (p = 0.0692). There was an absolute 26% reduction in mortality for the combined HBO2/NBH group (p = 0.048) and an absolute 36% improvement in favorable outcome using the sliding dichotomized GOS (p = 0.024) as compared with the control group. CONCLUSIONS In this Phase II clinical trial, in comparison with standard care (control treatment) combined HBO2/NBH treatments significantly improved markers of oxidative metabolism in relatively uninjured brain as well as pericontusional tissue, reduced intracranial hypertension, and demonstrated improvement in markers of cerebral toxicity. There was significant reduction in mortality and improved favorable outcome as measured by GOS. The combination of HBO2 and NBH therapy appears to have potential therapeutic efficacy as compared with the 2 treatments in isolation. CLINICAL TRIAL REGISTRATION NO.: NCT00170352 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Sarah B Rockswold
- Department of Physical Medicine and Rehabilitation, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | | | |
Collapse
|
99
|
Nicholson JD, Puche AC, Guo Y, Weinreich D, Slater BJ, Bernstein SL. PGJ(2) provides prolonged CNS stroke protection by reducing white matter edema. PLoS One 2012; 7:e50021. [PMID: 23284631 PMCID: PMC3527449 DOI: 10.1371/journal.pone.0050021] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2012] [Accepted: 10/17/2012] [Indexed: 11/18/2022] Open
Abstract
Few clinically effective approaches reduce CNS-white matter injury. After early in-vivo white matter infarct, NFκB-driven pro-inflammatory signals can amplify a relatively small amount of vascular damage, resulting in progressive endothelial dysfunction to create a severe ischemic lesion. This process can be minimized by 15-deoxy-Δ(12,14)-prostaglandin J2 (PGJ(2)), an analog of the metabolically active PGD(2) metabolite. We evaluated PGJ(2)'s effects and mechanisms using rodent anterior ischemic optic neuropathy (rAION); an in vivo white matter ischemia model. PGJ(2) administration systemically administered either acutely or 5 hours post-insult results in significant neuroprotection, with stereologic evaluation showing improved neuronal survival 30 days post-infarct. Quantitative capillary vascular analysis reveals that PGJ(2) improves perfusion at 1 day post-infarct by reducing tissue edema. Our results suggest that PGJ(2) acts by reducing NFκB signaling through preventing p65 nuclear localization and inhibiting inflammatory gene expression. Importantly, PGJ(2) showed no in vivo toxicity structurally as measured by optic nerve (ON) myelin thickness, functionally by ON-compound action potentials, on a cellular basis by oligodendrocyte precursor survival or changes in ON-myelin gene expression. PGJ(2) may be a clinically useful neuroprotective agent for ON and other CNS infarcts involving white matter, with mechanisms of action enabling effective treatment beyond the currently considered maximal time for intervention.
Collapse
Affiliation(s)
- James D. Nicholson
- Department of Ophthalmology and Visual Sciences, University of Maryland-Baltimore School of Medicine, Baltimore, Maryland, United States of America
| | - Adam C. Puche
- Department of Anatomy and Neurobiology, University of Maryland-Baltimore School of Medicine, Baltimore, Maryland, United States of America
| | - Yan Guo
- Department of Ophthalmology and Visual Sciences, University of Maryland-Baltimore School of Medicine, Baltimore, Maryland, United States of America
| | - Daniel Weinreich
- Department of Pharmacology, University of Maryland-Baltimore School of Medicine, Baltimore, Maryland, United States of America
| | - Bernard J. Slater
- Department of Ophthalmology and Visual Sciences, University of Maryland-Baltimore School of Medicine, Baltimore, Maryland, United States of America
| | - Steven L. Bernstein
- Department of Ophthalmology and Visual Sciences, University of Maryland-Baltimore School of Medicine, Baltimore, Maryland, United States of America
- Department of Anatomy and Neurobiology, University of Maryland-Baltimore School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
100
|
Kahle KT, Walcott BP, Simard JM. Continuous hyperosmolar therapy for traumatic brain injury-associated cerebral edema: as good as it gets, or an iatrogenic secondary insult? J Clin Neurosci 2012. [PMID: 23200533 DOI: 10.1016/j.jocn.2012.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Cerebral edema is a heterogeneous condition that is present in almost every type of neurological disease process--ranging from tumor, to cerebrovascular disease, to infection, to trauma, among others. It is associated with a high rate of morbidity and mortality. The pathophysiologic mechanisms of edema formation are distinct for the different conditions, thereby defining the various classifications. A relatively new treatment practice for cerebral edema is known as induced, sustained hypernatremia. This practice is highly controversial, is in widespread use, and lacks robust evidence for efficacy. Herein, we review details of the controversy regarding this practice.
Collapse
Affiliation(s)
- Kristopher T Kahle
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | |
Collapse
|