Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Cigdem Tosun is active.

Publication


Featured researches published by Cigdem Tosun.


Journal of Cerebral Blood Flow and Metabolism | 2009

Glibenclamide reduces inflammation, vasogenic edema, and caspase-3 activation after subarachnoid hemorrhage

J. Marc Simard; Zhihua Geng; S. Kyoon Woo; Svetlana Ivanova; Cigdem Tosun; Ludmila Melnichenko; Volodymyr Gerzanich

Subarachnoid hemorrhage (SAH) causes secondary brain injury due to vasospasm and inflammation. Here, we studied a rat model of mild-to-moderate SAH intended to minimize ischemia/hypoxia to examine the role of sulfonylurea receptor 1 (SUR1) in the inflammatory response induced by SAH. mRNA for Abcc8, which encodes SUR1, and SUR1 protein were abundantly upregulated in cortex adjacent to SAH, where tumor-necrosis factor-α (TNFα) and nuclear factor (NF)κB signaling were prominent. In vitro experiments confirmed that Abcc8 transcription is stimulated by TNFα. To investigate the functional consequences of SUR1 expression after SAH, we studied the effect of the potent, selective SUR1 inhibitor, glibenclamide. We examined barrier permeability (immunoglobulin G, IgG extravasation), and its correlate, the localization of the tight junction protein, zona occludens 1 (ZO-1). SAH caused a large increase in barrier permeability and disrupted the normal junctional localization of ZO-1, with glibenclamide significantly reducing both effects. In addition, SAH caused large increases in markers of inflammation, including TNFα and NFκB, and markers of cell injury or cell death, including IgG endocytosis and caspase-3 activation, with glibenclamide significantly reducing these effects. We conclude that block of SUR1 by glibenclamide may ameliorate several pathologic effects associated with inflammation that lead to cortical dysfunction after SAH.


Journal of Neurotrauma | 2011

Rodent model of direct cranial blast injury.

Reed Kuehn; Philippe F. Simard; Ian R. Driscoll; Kaspar Keledjian; Svetlana Ivanova; Cigdem Tosun; Alicia Williams; Grant V. Bochicchio; Volodymyr Gerzanich; J. Marc Simard

Traumatic brain injury resulting from an explosive blast is one of the most serious wounds suffered by warfighters, yet the effects of explosive blast overpressure directly impacting the head are poorly understood. We developed a rodent model of direct cranial blast injury (dcBI), in which a blast overpressure could be delivered exclusively to the head, precluding indirect brain injury via thoracic transmission of the blast wave. We constructed and validated a Cranium Only Blast Injury Apparatus (COBIA) to deliver blast overpressures generated by detonating .22 caliber cartridges of smokeless powder. Blast waveforms generated by COBIA replicated those recorded within armored vehicles penetrated by munitions. Lethal dcBI (LD(50) ∼ 515 kPa) was associated with: (1) apparent brainstem failure, characterized by immediate opisthotonus and apnea leading to cardiac arrest that could not be overcome by cardiopulmonary resuscitation; (2) widespread subarachnoid hemorrhages without cortical contusions or intracerebral or intraventricular hemorrhages; and (3) no pulmonary abnormalities. Sub-lethal dcBI was associated with: (1) apnea lasting up to 15 sec, with transient abnormalities in oxygen saturation; (2) very few delayed deaths; (3) subarachnoid hemorrhages, especially in the path of the blast wave; (4) abnormal immunolabeling for IgG, cleaved caspase-3, and β-amyloid precursor protein (β-APP), and staining for Fluoro-Jade C, all in deep brain regions away from the subarachnoid hemorrhages, but in the path of the blast wave; and (5) abnormalities on the accelerating Rotarod that persisted for the 1 week period of observation. We conclude that exposure of the head alone to severe explosive blast predisposes to significant neurological dysfunction.


Journal of Neurotrauma | 2009

Novel Model of Frontal Impact Closed Head Injury in the Rat

Michael Kilbourne; Reed Kuehn; Cigdem Tosun; John Caridi; Kaspar Keledjian; Grant V. Bochicchio; Thomas M. Scalea; Volodymyr Gerzanich; J. Marc Simard

Frontal impact, closed head trauma is a frequent cause of traumatic brain injury (TBI) in motor vehicle and sports accidents. Diffuse axonal injury (DAI) is common in humans and experimental animals, and results from shearing forces that develop within the anisotropic brain. Because the specific anisotropic properties of the brain are axis-dependent, the anatomical site where force is applied as well as the resultant acceleration, be it linear, rotational, or some combination, are important determinants of the resulting pattern of brain injury. Available rodent models of closed head injury do not reproduce the frontal impact commonly encountered in humans. Here we describe a new rat model of closed head injury that is a modification of the impact-acceleration model of Marmarou. In our model (the Maryland model), the impact force is applied to the anterior part of the cranium and produces TBI by causing anterior-posterior plus sagittal rotational acceleration of the brain inside the intact cranium. Skull fractures, prolonged apnea, and mortality were absent. The animals exhibited petechial hemorrhages, DAI marked by a bead-like pattern of beta-amyloid precursor protein (beta-APP) in damaged axons, and widespread upregulation of beta-APP in neurons, with regions affected including the orbitofrontal cortex (coup), corpus callosum, caudate, putamen, thalamus, cerebellum, and brainstem. Activated caspase-3 was prominent in hippocampal neurons and Purkinje cells at the grey-white matter junction of the cerebellum. Neurobehavioral dysfunction, manifesting as reduced spontaneous exploration, lasted more than 1 week. We conclude that the Maryland model produces diffuse injuries that may be relevant to human brain injury.


Science Translational Medicine | 2010

Brief Suppression of Abcc8 Prevents Autodestruction of Spinal Cord After Trauma

J. Marc Simard; S. Kyoon Woo; Michael D. Norenberg; Cigdem Tosun; Zheng Chen; Svetlana Ivanova; Orest Tsymbalyuk; Joseph Bryan; Douglas Landsman; Volodymyr Gerzanich

Secondary injury that occurs after trauma to the spinal cord can be prevented by inhibiting expression of the gene that regulates a cation transporter. Tackling Spinal Cord Injury Damage to the brain has a way of spreading. The initial injury often sparks a secondary wave of destruction that enlarges the damaged area and increases the ultimate disability of the patient. This process presents a tempting target for therapeutic intervention and, indeed, numerous agents interfere with secondary injury in brain-damaged animals. But none of these potential drugs have proved effective in humans. Simard and his colleagues now hope to bypass these previous dead ends and successfully interfere with secondary damage by basing their animal work on data taken from human victims of spinal cord injury. These authors examined brain tissue from seven patients who had died shortly after traumatic injury to the spinal cord and show that one prominent sequel of local damage is that the surrounding tissues show higher than normal concentrations of messenger RNA (mRNA) and protein for the sulfonylurea receptor 1 (SUR1). Their data from rat and mouse show the same thing. This receptor associates with pores in cell membranes to form ion channels, one of which causes cell depolarization and ultimately cell death, creating the wave of secondary damage to the cord. Simard et al. then report that mice in which SUR1 had been genetically removed suffer much less damage to the spinal cord after injury, a result of a less robust wave of spreading damage. Treatment of rats, a better model of human spinal cord injury than mice, with antisense nucleotides that inhibit SUR1 mRNA or with glibenclamide, a nonspecific inhibitor of the whole class of SUR-like proteins, both protected against secondary injury. The capillaries in the cord surrounding the injury were intact rather than fragmented as they are in untreated rats, and the treated rats performed better on a battery of behavioral tests, showing their superior neurological function. Upon later examination, the size of the lesion in the treated animals was only one-quarter the size of the lesion in control animals. SUR1, therefore, may be a critical element in causing the secondary damage of brain trauma, in humans and rodents. Therapeutic agents that interfere with its injury-induced stimulation of ion channels should be tested in injured patients to determine whether the devastating disability that often results from spinal cord injury can be minimized. Spinal cord injury (SCI) is typically complicated by progressive hemorrhagic necrosis, an autodestructive process of secondary injury characterized by progressive enlargement of a hemorrhagic contusion during the first several hours after trauma. We assessed the role of Abcc8, which encodes sulfonylurea receptor 1 (SUR1), in progressive hemorrhagic necrosis. After SCI, humans and rodents exhibited similar regional and cellular patterns of up-regulation of SUR1 and Abcc8 messenger RNA. Elimination of SUR1 in Abcc8−/− mice and in rats given antisense oligodeoxynucleotide against Abcc8 prevented progressive hemorrhagic necrosis, yielded significantly better neurological function, and resulted in lesions that were one-fourth to one-third the size of those in control animals. The beneficial effects of Abcc8 suppression were associated with prevention of oncotic (necrotic) death of capillary endothelial cells. Suppression of Abcc8 with antisense oligodeoxynucleotide after SCI presents an opportunity for reducing the devastating sequelae of SCI.


Stroke | 2013

Inhibition of the Sur1-Trpm4 Channel Reduces Neuroinflammation and Cognitive Impairment in Subarachnoid Hemorrhage

Cigdem Tosun; David B. Kurland; Rupal I. Mehta; Rudy J. Castellani; Joyce L. deJong; Min Seong Kwon; Seung Kyoon Woo; Volodymyr Gerzanich; J. Marc Simard

Background and Purpose— Subarachnoid hemorrhage (SAH) can leave patients with memory impairments that may not recover fully. Molecular mechanisms are poorly understood, and no treatment is available. The sulfonylurea receptor 1–transient receptor potential melastatin 4 (Sur1-Trpm4) channel plays an important role in acute central nervous system injury. We evaluated upregulation of Sur1-Trpm4 in humans with SAH and, in rat models of SAH, we examined Sur1-Trpm4 upregulation, its role in barrier dysfunction and neuroinflammation, and its consequences on spatial learning. Methods— We used Förster resonance energy transfer to detect coassociated Sur1 and Trpm4 in human autopsy brains with SAH. We studied rat models of SAH involving filament puncture of the internal carotid artery or injection of blood into the subarachnoid space of the entorhinal cortex. In rats, we used Förster resonance energy transfer and coimmunoprecipitation to detect coassociated Sur1 and Trpm4, we measured immunoglobulin G extravasation and tumor necrosis &agr; overexpression as measures of barrier dysfunction and neuroinflammation, and we assessed spatial learning and memory on days 7 to 19. Results— Sur1-Trpm4 channels were upregulated in humans and rats with SAH. In rats, inhibiting Sur1 using antisense or the selective Sur1 inhibitor glibenclamide reduced SAH-induced immunoglobulin G extravasation and tumor necrosis &agr; overexpression. In models with entorhinal SAH, rats treated with glibenclamide for 7 days after SAH exhibited better platform search strategies and better performance on incremental and rapid spatial learning than vehicle-treated controls. Conclusions— Sur1-Trpm4 channels are upregulated in humans and rats with SAH. Channel inhibition with glibenclamide may reduce neuroinflammation and the severity of cognitive deficits after SAH.


Brain Sciences | 2013

The Protective Effect of Glibenclamide in a Model of Hemorrhagic Encephalopathy of Prematurity

Cigdem Tosun; Michael T. Koltz; David B. Kurland; Hina Ijaz; Melda Gurakar; Gary Schwartzbauer; Turhan Coksaygan; Svetlana Ivanova; Volodymyr Gerzanich; J. Marc Simard

We studied a model of hemorrhagic encephalopathy of prematurity (EP) that closely recapitulates findings in humans with hemorrhagic EP. This model involves tandem insults of 20 min intrauterine ischemia (IUI) plus an episode of elevated venous pressure induced by intraperitoneal glycerol on post-natal day (P) 0. We examined Sur1 expression, which is upregulated after focal ischemia but has not been studied after brief global ischemia including IUI. We found that 20 min IUI resulted in robust upregulation of Sur1 in periventricular microvessels and tissues. We studied tandem insult pups from untreated or vehicle-treated dams (TI-CTR), and tandem insult pups from dams administered a low-dose, non-hypoglycemogenic infusion of the Sur1 blocker, glibenclamide, for 1 week after IUI (TI-GLIB). Compared to pups from the TI-CTR group, pups from the TI-GLIB group had significantly fewer and less severe hemorrhages on P1, performed significantly better on the beam walk and accelerating Rotarod on P35 and in tests of thigmotaxis and rapid learning on P35–49, and had significantly greater body and brain weights at P52. We conclude that low-dose glibenclamide administered to the mother at the end of pregnancy protects pups subjected to IUI from post-natal events of elevated venous pressure and its consequences.


Journal of Neuropathology and Experimental Neurology | 2013

Sulfonylurea receptor 1 expression in human cerebral infarcts.

Rupal I. Mehta; Svetlana Ivanova; Cigdem Tosun; Rudy J. Castellani; Volodymyr Gerzanich; J. Marc Simard

Supplemental Digital Content is available in the article.


Journal of Neurotrauma | 2014

Exposure of the Thorax to a Sublethal Blast Wave Causes a Hydrodynamic Pulse That Leads to Perivenular Inflammation in the Brain

Simard Jm; Pampori A; Kaspar Keledjian; Cigdem Tosun; Schwartzbauer G; Svetlana Ivanova; Gerzanich

Traumatic brain injury (TBI) caused by an explosive blast (blast-TBI) is postulated to result, in part, from transvascular transmission to the brain of a hydrodynamic pulse (a.k.a., volumetric blood surge, ballistic pressure wave, hydrostatic shock, or hydraulic shock) induced in major intrathoracic blood vessels. This mechanism of blast-TBI has not been demonstrated directly. We tested the hypothesis that a blast wave impacting the thorax would induce a hydrodynamic pulse that would cause pathological changes in the brain. We constructed a Thorax-Only Blast Injury Apparatus (TOBIA) and a Jugular-Only Blast Injury Apparatus (JOBIA). TOBIA delivered a collimated blast wave to the right lateral thorax of a rat, precluding direct impact on the cranium. JOBIA delivered a blast wave to the fluid-filled port of an extracorporeal intravenous infusion device whose catheter was inserted retrograde into the jugular vein, precluding lung injury. Long Evans rats were subjected to sublethal injury by TOBIA or JOBIA. Blast injury induced by TOBIA was characterized by apnea and diffuse bilateral hemorrhagic injury to the lungs associated with a transient reduction in pulse oximetry signals. Immunolabeling 24 h after injury by TOBIA showed up-regulation of tumor necrosis factor alpha, ED-1, sulfonylurea receptor 1 (Sur1), and glial fibrillary acidic protein in veins or perivenular tissues and microvessels throughout the brain. The perivenular inflammatory effects induced by TOBIA were prevented by ligating the jugular vein and were reproduced using JOBIA. We conclude that blast injury to the thorax leads to perivenular inflammation, Sur1 up-regulation, and reactive astrocytosis resulting from the induction of a hydrodynamic pulse in the vasculature.


Biomarkers | 2014

Biomarkers as outcome predictors in subarachnoid hemorrhage – a systematic review

Caron M. Hong; Cigdem Tosun; David B. Kurland; Volodymyr Gerzanich; David Schreibman; J. Marc Simard

Abstract Context: Subarachnoid hemorrhage (SAH) has a high fatality rate and many suffer from delayed neurological deficits. Biomarkers may aid in the identification of high-risk patients, guide treatment/management and improve outcome. Objective: The aim of this review was to summarize biomarkers of SAH associated with outcome. Methods: An electronic database query was completed, including an additional review of reference lists to include all potential human studies. Results: A total of 298 articles were identified; 112 were reviewed; 55 studies were included. Conclusion: This review details biomarkers of SAH that correlate with outcome. It provides the basis for research investigating their possible translation into the management of SAH patients.


Journal of Neuropathology and Experimental Neurology | 2015

Sur1-Trpm4 Cation Channel Expression in Human Cerebral Infarcts

Rupal I. Mehta; Cigdem Tosun; Svetlana Ivanova; Natalia Tsymbalyuk; Bolanle M. Famakin; Min Seong Kwon; Rudy J. Castellani; Volodymyr Gerzanich; J. Marc Simard

Abstract The nonselective monovalent cation channel transient receptor potential melastatin 4 (Trpm4) is transcriptionally upregulated in neural and vascular cells in animal models of brain infarction. It associates with sulfonylurea receptor 1 (Sur1) to form Sur1-Trpm4 channels, which have critical roles in cytotoxic edema, cell death, blood-brain barrier breakdown, and vasogenic edema. We examined Trpm4 expression in postmortem brain specimens from 15 patients who died within the first 31 days of the onset of focal cerebral ischemia. We found increased Trpm4 protein expression in all cases using immunohistochemistry; transcriptional upregulation was confirmed using in situ hybridization of Trpm4 messenger RNA. Transient receptor potential melastatin 4 colocalized and coassociated with Sur1 within ischemic endothelial cells and neurons. Coexpression of Sur1 and Trpm4 in necrotic endothelial cells was also associated with vasogenic edema indicated by upregulated perivascular tumor necrosis factor, extravasation of serum immunoglobulin G, and associated inflammation. Upregulated Trpm4 protein was present up to 1 month after the onset of cerebral ischemia. In a rat model of middle cerebral artery occlusion stroke, pharmacologic channel blockade by glibenclamide, a selective inhibitor of sulfonylurea receptor, mitigated perivascular tumor necrosis factor labeling. Thus, upregulated Sur1-Trpm4 channels and associated blood-brain barrier disruption and cerebral edema suggest that pharmacologic targeting of this channel may represent a promising therapeutic strategy for the clinical management of patients with cerebral ischemia.

Collaboration


Dive into the Cigdem Tosun's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Grant V. Bochicchio

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge