Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Glia ; 62(1): 26-38, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24166800

ABSTRACT

Traumatic brain injury (TBI) is a major cause of mortality and morbidity worldwide. Cerebral edema, a life-threatening medical complication, contributes to elevated intracranial pressure (ICP) and a poor clinical prognosis after TBI. Unfortunately, treatment options to reduce post-traumatic edema remain suboptimal, due in part, to a dearth of viable therapeutic targets. Herein, we tested the hypothesis that cerebral innate immune responses contribute to edema development after TBI. Our results demonstrate that high-mobility group box protein 1 (HMGB1) was released from necrotic neurons via a NR2B-mediated mechanism. HMGB1 was clinically associated with elevated ICP in patients and functionally promoted cerebral edema after TBI in mice. The detrimental effects of HMGB1 were mediated, at least in part, via activation of microglial toll-like receptor 4 (TLR4) and the subsequent expression of the astrocytic water channel, aquaporin-4 (AQP4). Genetic or pharmacological (VGX-1027) TLR4 inhibition attenuated the neuroinflammatory response and limited post-traumatic edema with a delayed, clinically implementable therapeutic window. Human and rodent tissue culture studies further defined the cellular mechanisms demonstrating neuronal HMGB1 initiates the microglial release of interleukin-6 (IL-6) in a TLR4 dependent mechanism. In turn, microglial IL-6 increased the astrocytic expression of AQP4. Taken together, these data implicate microglia as key mediators of post-traumatic brain edema and suggest HMGB1-TLR4 signaling promotes neurovascular dysfunction after TBI.


Subject(s)
Brain Edema/etiology , Brain Injuries/complications , HMGB1 Protein/metabolism , Microglia/metabolism , Neurons/metabolism , Toll-Like Receptor 4/metabolism , Acetates/pharmacology , Animals , Brain Edema/pathology , Brain Injuries/cerebrospinal fluid , Cells, Cultured , Cerebral Cortex/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Humans , Immunologic Factors/pharmacology , Male , Mice , Mice, Inbred C3H , Microglia/drug effects , Neurons/drug effects , Oxazoles/pharmacology , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Toll-Like Receptor 4/genetics
2.
Transl Stroke Res ; 4(6): 643-51, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24323418

ABSTRACT

Mitochondrial respiratory capacity is critical for responding to changes in neuronal energy demand. One approach toward neuroprotection is the administration of alternative energy substrates ("biofuels") to overcome brain injury-induced inhibition of glucose-based aerobic energy metabolism. This study tested the hypothesis that exogenous pyruvate, lactate, ß-hydroxybutyrate, and acetyl-L-carnitine each increase neuronal respiratory capacity in vitro either in the absence of or following transient excitotoxic glutamate receptor stimulation. Compared to the presence of 5 mM glucose alone, the addition of pyruvate, lactate, or ß-hydroxybutyrate (1.0-10.0 mM) to either day in vitro (DIV) 14 or 7 rat cortical neurons resulted in significant, dose-dependent stimulation of respiratory capacity, measured by cell respirometry as the maximal O2 consumption rate in the presence of the respiratory uncoupler carbonyl cyanide-p-trifluoromethoxyphenylhydrazone. A 30-min exposure to 100 µM glutamate impaired respiratory capacity for DIV 14, but not DIV 7, neurons. Glutamate reduced the respiratory capacity for DIV 14 neurons with glucose alone by 25 % and also reduced respiratory capacity with glucose plus pyruvate, lactate, or ß-hydroxybutyrate. However, respiratory capacity in glutamate-exposed neurons following pyruvate or ß-hydroxybutyrate addition was still, at least, as high as that obtained with glucose alone in the absence of glutamate exposure. These results support the interpretation that previously observed neuroprotection by exogenous pyruvate, lactate, or ß-hydroxybutyrate is at least partially mediated by their preservation of neuronal respiratory capacity.


Subject(s)
Cerebral Cortex/drug effects , Glutamic Acid/pharmacology , Mitochondria/drug effects , Neurons/drug effects , Oxygen Consumption/drug effects , 3-Hydroxybutyric Acid/pharmacology , Acetylcarnitine/pharmacology , Animals , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cell Respiration/drug effects , Cerebral Cortex/metabolism , Dose-Response Relationship, Drug , Energy Metabolism/drug effects , Glutamic Acid/metabolism , In Vitro Techniques , Lactic Acid/pharmacology , Mitochondria/metabolism , Neurons/metabolism , Nootropic Agents/pharmacology , Proton Ionophores/pharmacology , Pyruvic Acid/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Glutamate/metabolism
3.
PLoS One ; 7(4): e34504, 2012.
Article in English | MEDLINE | ID: mdl-22485176

ABSTRACT

BACKGROUND: Oxidative stress is known to play an important role in the pathology of traumatic brain injury. Mitochondria are thought to be the major source of the damaging reactive oxygen species (ROS) following TBI. However, recent work has revealed that the membrane, via the enzyme NADPH oxidase can also generate the superoxide radical (O(2)(-)), and thereby potentially contribute to the oxidative stress following TBI. The current study thus addressed the potential role of NADPH oxidase in TBI. METHODOLOGY/PRINCIPAL FINDINGS: The results revealed that NADPH oxidase activity in the cerebral cortex and hippocampal CA1 region increases rapidly following controlled cortical impact in male mice, with an early peak at 1 h, followed by a secondary peak from 24-96 h after TBI. In situ localization using oxidized hydroethidine and the neuronal marker, NeuN, revealed that the O(2)(-) induction occurred in neurons at 1 h after TBI. Pre- or post-treatment with the NADPH oxidase inhibitor, apocynin markedly inhibited microglial activation and oxidative stress damage. Apocynin also attenuated TBI-induction of the Alzheimer's disease proteins ß-amyloid and amyloid precursor protein. Finally, both pre- and post-treatment of apocynin was also shown to induce significant neuroprotection against TBI. In addition, a NOX2-specific inhibitor, gp91ds-tat was also shown to exert neuroprotection against TBI. CONCLUSIONS/SIGNIFICANCE: As a whole, the study demonstrates that NADPH oxidase activity and superoxide production exhibit a biphasic elevation in the hippocampus and cortex following TBI, which contributes significantly to the pathology of TBI via mediation of oxidative stress damage, microglial activation, and AD protein induction in the brain following TBI.


Subject(s)
Brain Injuries/enzymology , Membrane Glycoproteins/metabolism , Microglia/physiology , NADPH Oxidases/metabolism , Neurons/enzymology , Acetophenones/pharmacology , Amyloid beta-Peptides/metabolism , Animals , Brain Edema/enzymology , Brain Edema/pathology , Brain Injuries/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Enzyme Activation , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Male , Membrane Glycoproteins/antagonists & inhibitors , Mice , Microglia/drug effects , Microglia/metabolism , NADPH Oxidase 2 , NADPH Oxidases/antagonists & inhibitors , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Oxidation-Reduction , Oxidative Stress , Superoxides/metabolism
4.
Int J Dev Neurosci ; 29(7): 701-10, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21704149

ABSTRACT

Neuroblastoma (NB) is the most prevalent pediatric solid tumor and a leading cause of cancer-related death in children. In the present study, a novel cytotoxic role for the dietary compounds, curcumin, andrographolide, wedelolactone, dibenzoylmethane, and tanshinone IIA was identified in human S-type NB cells, SK-N-AS and SK-N-BE(2). Mechanistically, cell death appeared apoptotic by flow cytometry; however, these effects proceeded independently from both caspase-3 and p53 activation, as assessed by both genetic (shRNA) and pharmacological approaches. Notably, cell death induced by both curcumin and andrographolide was associated with decreased NFκB activity and a reduction in Bcl-2 and Bcl-xL expression. Finally, curcumin and andrographolide increased cytotoxicity following co-treatment with either cisplatin or doxorubicin, two chemotherapeutic agents widely used in the clinical management of NB. Coupled with the documented safety in humans, dietary compounds may represent a potential adjunct therapy for NB.


Subject(s)
Antineoplastic Agents , Caspases/metabolism , Cell Death/drug effects , Diet , Neuroblastoma , Plant Extracts/chemistry , Tumor Suppressor Protein p53/metabolism , Abietanes/administration & dosage , Abietanes/pharmacology , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Chalcones/administration & dosage , Chalcones/pharmacology , Chromones/pharmacology , Coumarins/administration & dosage , Coumarins/pharmacology , Curcumin/administration & dosage , Curcumin/pharmacology , Diterpenes/administration & dosage , Diterpenes/pharmacology , Humans , Morpholines/pharmacology , NF-kappa B/metabolism , Neuroblastoma/diet therapy , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/genetics , bcl-X Protein/genetics , bcl-X Protein/metabolism
5.
Glia ; 58(15): 1858-70, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20737478

ABSTRACT

Intracerebral hemorrhage (ICH) induces neurovascular injury via poorly defined mechanisms. The aim of this study was to determine whether gliovascular communication may restrict hemorrhagic vascular injury. Hemin, a hemoglobin by-product, concentration- and time-dependently increased apoptotic cell death in mouse bEnd.3 cells and in primary human brain microvascular endothelial cells, at least in part, via a caspase-3 dependent pathway. Cell death was preceded by a NFκB-mediated increase in inflammatory gene expression, including upregulation of inducible nitric oxide synthase (iNOS) expression and activity. Functionally, inhibition of iNOS or the addition of a peroxynitrite decomposition catalyst reduced cell death. Interestingly, co-treatment with astrocyte-conditioned media (ACM) reversed hemin-induced NFκB activation, nitrotyrosine formation, and apoptotic cell death, at least in part, via the release of the endogenous antioxidant, reduced glutathione (GSH). Prior treatment of astrocytes with the GSH-depleting agent, DL-buthionine (S,R)-sulfoximine or direct addition of diethyl maleate, a thiol-depleting agent, to ACM reversed the observed protection. In contrast, neither exogenous GSH nor the GSH precursor, N-acetylcysteine, was protective in bEnd.3 cells. Together, these data support an important role for astrocyte-derived GSH in the maintenance of oxidative balance in the vasculature and suggest therapeutic targeting of the GSH system may reduce neurological injury following ICH.


Subject(s)
Apoptosis/drug effects , Astrocytes/chemistry , Glutathione/pharmacology , Hemin/pharmacology , Microvessels/cytology , Acetylcysteine/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Brain/cytology , Caspase 3/metabolism , Cells, Cultured , Curcumin/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Glutathione/metabolism , Humans , L-Lactate Dehydrogenase/metabolism , Mice , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type II/metabolism , Peroxynitrous Acid/pharmacology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Time Factors
6.
J Neurosurg ; 113(6): 1195-201, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20397893

ABSTRACT

OBJECT: Traumatic brain injury (TBI) induces significant neurological damage, including deficits in learning and memory, which contribute to a poor clinical prognosis. Treatment options to limit cognitive decline and promote neurological recovery are lacking, in part due to a poor understanding of the secondary or delayed processes that contribute to brain injury. In the present study, the authors characterized the temporal and spatial changes in the expression of postsynaptic density protein-95 (PSD-95), a key scaffolding protein implicated in excitatory synaptic signaling, after controlled cortical impacts in mice. Neurological injury, as assessed by the open-field activity test and the novel object recognition test, was compared with changes in PSD-95 expression. METHODS: Adult male CD-1 mice were subjected to controlled cortical impacts to simulate moderate TBI in humans. The spatial and temporal expression of PSD-95 was analyzed in the cerebral cortex and hippocampus at various time points following injury and sham operations. Neurological assessments were performed to compare changes in PSD-95 with cognitive deficits. RESULTS: A significant decrease in PSD-95 expression was observed in the ipsilateral hippocampus beginning on Day 7 postinjury. The loss of PSD-95 corresponded with a concomitant reduction in immunoreactivity for NeuN (neuronal nuclei), a neuron-specific marker. Aside from the contused cortex, a significant loss of PSD-95 immunoreactivity was not observed in the cerebral cortex. The delayed loss of hippocampal PSD-95 directly correlated with the onset of behavioral deficits, suggesting a possible causative role for PSD-95 in behavioral abnormalities following head trauma. CONCLUSIONS: A delayed loss of hippocampal synapses was observed following head trauma in mice. These data may suggest a cellular mechanism to explain the delayed learning and memory deficits in humans after TBI and provide a potential framework for further testing to implicate PSD-95 as a clinically relevant therapeutic target.


Subject(s)
Brain Injuries/complications , Cerebral Cortex/injuries , Cognition Disorders/metabolism , Hippocampus/metabolism , Membrane Proteins/biosynthesis , Animals , Brain Injuries/metabolism , Cerebral Cortex/metabolism , Cognition Disorders/etiology , Disease Models, Animal , Disks Large Homolog 4 Protein , Guanylate Kinases , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred Strains , Time Factors
7.
J Neurochem ; 113(3): 637-48, 2010 May.
Article in English | MEDLINE | ID: mdl-20132469

ABSTRACT

Traumatic brain injury is a devastating neurological injury associated with significant morbidity and mortality. Medical therapies to limit cerebral edema, a cause of increased intracranial hypertension and poor clinical outcome, are largely ineffective, emphasizing the need for novel therapeutic approaches. In the present study, pre-treatment with curcumin (75, 150 mg/kg) or 30 min post-treatment with 300 mg/kg significantly reduced brain water content and improved neurological outcome following a moderate controlled cortical impact in mice. The protective effect of curcumin was associated with a significant attenuation in the acute pericontusional expression of interleukin-1beta, a pro-inflammatory cytokine, after injury. Curcumin also reversed the induction of aquaporin-4, an astrocytic water channel implicated in the development of cellular edema following head trauma. Notably, curcumin blocked IL-1beta-induced aquaporin-4 expression in cultured astrocytes, an effect mediated, at least in part, by reduced activation of the p50 and p65 subunits of nuclear factor kappaB. Consistent with this notion, curcumin preferentially attenuated phosphorylated p65 immunoreactivity in pericontusional astrocytes and decreased the expression of glial fibrillary acidic protein, a reactive astrocyte marker. As a whole, these data suggest clinically achievable concentrations of curcumin reduce glial activation and cerebral edema following neurotrauma, a finding which warrants further investigation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aquaporin 4/antagonists & inhibitors , Brain Edema/etiology , Brain Edema/prevention & control , Brain Injuries/complications , Curcumin/pharmacology , Animals , Blotting, Western , Brain Edema/pathology , Brain Injuries/pathology , Brain Injuries/psychology , Cells, Cultured , Immunohistochemistry , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/biosynthesis , Interleukin-1beta/physiology , Learning/physiology , Male , Memory/physiology , Mice , Microscopy, Confocal , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Recognition, Psychology/physiology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
8.
Neurosurg Focus ; 28(1): E10, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20043714

ABSTRACT

Subarachnoid hemorrhage (SAH) is a devastating neurological injury associated with significant patient morbidity and death. Since the first demonstration of cerebral vasospasm nearly 60 years ago, the preponderance of research has focused on strategies to limit arterial narrowing and delayed cerebral ischemia following SAH. However, recent clinical and preclinical data indicate a functional dissociation between cerebral vasospasm and neurological outcome, signaling the need for a paradigm shift in the study of brain injury following SAH. Early brain injury may contribute to poor outcome and early death following SAH. However, elucidation of the complex cellular mechanisms underlying early brain injury remains a major challenge. The advent of modern neuroproteomics has rapidly advanced scientific discovery by allowing proteome-wide screening in an objective, nonbiased manner, providing novel mechanisms of brain physiology and injury. In the context of neurosurgery, proteomic analysis of patient-derived CSF will permit the identification of biomarkers and/or novel drug targets that may not be intuitively linked with any particular disease. In the present report, the authors discuss the utility of neuroproteomics with a focus on the roles for this technology in understanding SAH. The authors also provide data from our laboratory that identifies high-mobility group box protein-1 as a potential biomarker of neurological outcome following SAH in humans.


Subject(s)
Brain/physiopathology , Proteome/physiology , Proteomics/methods , Subarachnoid Hemorrhage/physiopathology , Biomarkers/analysis , Biomarkers/cerebrospinal fluid , Forecasting , HMGB1 Protein/genetics , HMGB1 Protein/physiology , Humans , Intracranial Aneurysm/cerebrospinal fluid , Intracranial Aneurysm/genetics , Intracranial Aneurysm/physiopathology , Neurosurgery , Proteomics/trends , Stroke/physiopathology , Subarachnoid Hemorrhage/cerebrospinal fluid , Subarachnoid Hemorrhage/surgery , Treatment Outcome , Vasospasm, Intracranial/physiopathology
9.
J Appl Physiol (1985) ; 106(2): 476-85, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19023026

ABSTRACT

A short recovery period between same-day competitions is common practice in organized youth sports. We hypothesized that young athletes will experience an increase in physiological strain and perceptual discomfort during a second identical exercise bout in the heat, with 1 h (21 degrees C) between bouts, even with ample hydration. Twenty-four athletes (6 boys and 6 girls: 12-13 yr old, 47.7 +/- 8.3 kg; 6 boys and 6 girls: 16-17 yr old, 61.0 +/- 8.6 kg) completed two 80-min intermittent exercise bouts (treadmill 60%, cycle 40% peak oxygen uptake) in the heat (33 degrees C, 48.9 +/- 6.1% relative humidity). Sweat loss during each bout was similar within each age group (12-13 yr old: bout 1, 943.6 +/- 237.1 ml; bout 2, 955.5 +/- 250.3 ml; 16-17 yr old: bout 1, 1,382.2 +/- 480.7 ml; bout 2, 1,373.1 +/- 472.2 ml). Area under the curve (AUC) was not statistically different (P > 0.05) between bouts for core body temperature (12-13 yr old: bout 1 peak, 38.6 +/- 0.4 degrees C; bout 2, 38.4 +/- 0.2 degrees C; 16-17 yr old: bout 1 peak, 38.8 +/- 0.7 degrees C; bout 2, 38.7 +/- 0.6 degrees C), physiological strain index (12-13 yr old: bout 1 peak, 7.9 +/- 0.9; bout 2, 7.5 +/- 0.7; 16-17 yr old: bout 1 peak, 8.1 +/- 1.5; bout 2, 7.9 +/- 1.4), or thermal sensation for any age/sex subgroup or for all subjects combined. However, rating of perceived exertion AUC and peak were higher (P = 0.0090 and 0.0004, respectively) during bout 2 in the older age group. Notably, four subjects experienced consistently higher responses throughout bout 2. With these healthy, fit, young athletes, 1 h of complete rest, cool down, and rehydration following 80 min of strenuous exercise in the heat was generally effective in eliminating any apparent carryover effects that would have resulted in greater thermal and cardiovascular strain during a subsequent identical exercise bout.


Subject(s)
Acclimatization , Exercise , Heat Stress Disorders/physiopathology , Hot Temperature , Perception , Adolescent , Age Factors , Body Temperature , Child , Female , Heart Rate , Heat Stress Disorders/prevention & control , Heat Stress Disorders/psychology , Humans , Male , Recovery of Function , Sweating , Time Factors , Water-Electrolyte Balance
10.
Antioxid Redox Signal ; 11(1): 35-45, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18752423

ABSTRACT

Cerebral vasospasm is a major cause of death and disability after subarachnoid hemorrhage (SAH); however, clinical therapies to limit the development of cerebral vasospasm are lacking. Although the causative factors underlying the development of cerebral vasospasm are poorly understood, oxidative stress contributes to disease progression. In the present study, curcumin (150 or 300 mg/kg) protected against the development of cerebral vasospasm and limited secondary cerebral infarction after SAH in mice. The protective effect of curcumin was associated with a significant attenuation of inflammatory gene expression and lipid peroxidation within the cerebral cortex and the middle cerebral artery. Despite the ability of curcumin to limit the development of cerebral vasospasm and secondary infarction, behavioral outcome was not improved, indicating a dissociation between cerebral vasospasm and neurologic outcome. Together, these data indicate a novel role for curcumin as a possible adjunct therapy after SAH, both to prevent the development of cerebral vasospasm and to reduce oxidative brain injury after secondary infarction.


Subject(s)
Curcumin/therapeutic use , Endothelium, Vascular/physiopathology , Inflammation/drug therapy , Subarachnoid Hemorrhage/complications , Vasospasm, Intracranial/drug therapy , Animals , Disease Models, Animal , Inflammation/complications , Inflammation/physiopathology , Lipid Peroxidation/drug effects , Male , Mice , Mice, Inbred Strains , NF-kappa B/metabolism , Subarachnoid Hemorrhage/chemically induced , Subarachnoid Hemorrhage/pathology , Superoxides/analysis , Superoxides/metabolism , Thiobarbituric Acid Reactive Substances/analysis , Vasospasm, Intracranial/complications
11.
Free Radic Biol Med ; 45(8): 1103-14, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18706498

ABSTRACT

Intracerebral hemorrhage (ICH) is a devastating neurological injury associated with significant mortality. Astrocytic inflammation may contribute to the pathogenesis of ICH, although the underlying cellular mechanisms remain unclear. In this study, the hemoglobin oxidation by-product, hemin, concentration dependently induced necroptotic cell death in cortical astrocytes within 5 h of treatment. Hemin-induced cell death was preceded by increased inflammatory gene expression (COX-2, IL-1beta, TNF-alpha, iNOS). Inhibition of the NF-kappaB transcription factor reversed inflammatory gene expression and attenuated cell death after hemin treatment, suggesting a possible role for inflammatory mediators in astrocytic injury. Superoxide production paralleled the increase in iNOS expression, and inhibition of either iNOS (aminoguanidine or iminopiperdine) or superoxide (apocynin) significantly reduced cell death. Similarly, reduced formation of peroxynitrite, the damaging product of nitric oxide and superoxide, significantly reduced hemin injury. Hemin-induced peroxidative injury was associated with a rapid depletion of intracellular glutathione (GSH), culminating in lipid peroxidation and cell death, effects that were reduced by cotreatment with exogenous GSH, N-acetyl-L-cysteine, or the glutathione peroxidase mimetic ebselen. Together, these studies suggest a novel role for GSH depletion in necroptotic astrocyte injury after a hemorrhagic injury and indicate that therapeutic targeting of GSH may exert a beneficial effect after ICH.


Subject(s)
Astrocytes/metabolism , Cell Death/physiology , Glutathione/metabolism , Hemin/metabolism , Animals , Astrocytes/pathology , Cells, Cultured , Gene Expression , Inflammation/metabolism , Lipid Peroxidation/physiology , Mice , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/physiology , Peroxynitrous Acid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Superoxides/metabolism
12.
Neurosurgery ; 62(5): 1122-7; discussion 1027-8, 2008 May.
Article in English | MEDLINE | ID: mdl-18580810

ABSTRACT

OBJECTIVE: Adenomas of the pituitary gland are among the most common types of tumors of the adult brain. Although adenomas are histologically benign, they may be associated with significant morbidity and mortality, mostly because of their invasive growth pattern and hormone hypersecretion. Current medical therapies are suppressive, acting at a receptor level. Thus, there is a need to identify novel cellular and molecular targets for pituitary tumors. We investigated the possible role of the NFkappaB transcription factor in pituitary tumor cell growth. METHODS: The effect of NFkappaB pathway inhibition on cellular viability was studied in the GH3 pituitary adenoma cell line, a well-characterized rat cell line that secretes growth hormone and prolactin. Cells were treated with mechanistically diverse pharmacological NFkappaB pathway inhibitors or with molecular inhibitors that were overexpressed in tumor cells before the assessment of cellular viability. NFkappaB activity was also assessed in GH3 cells using deoxyribonucleic acid binding assays. RESULTS: GH3 cells exhibited constitutive NFkappaB activity, which contributed to increased cellular proliferation. Treatment with wedelolactone, an IkappaB kinase inhibitor, or overexpression of an IkappaB super-repressor reduced cell viability, further implicating NFkappaB in pituitary tumor cell growth. Pharmacological or molecular inhibition of Akt similarly reduced GH3 viability and NFkappaB binding, suggesting that constitutive activation of NFkappaB may be, at least in part, mediated by Akt. CONCLUSION: Directed targeting of the Akt and NFkappaB signaling pathways may be a useful adjunct in the clinical management of pituitary tumors. Further elucidation of this pathway may yield novel information regarding the behavior of pituitary tumors in humans.


Subject(s)
NF-kappa B/metabolism , Pituitary Neoplasms/metabolism , Signal Transduction/physiology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Coumarins/pharmacology , Enzyme Inhibitors/pharmacology , NF-kappa B/drug effects , Proto-Oncogene Proteins c-akt , Rats
13.
Neurosignals ; 16(2-3): 154-64, 2008.
Article in English | MEDLINE | ID: mdl-18253055

ABSTRACT

Traumatic brain injury (TBI) is a leading cause of death and disability in the United States. Current medical therapies exhibit limited efficacy in reducing neurological injury and the prognosis for patients remains poor. While most research is focused on the direct protection of neuronal cells, non-neuronal cells, such as astrocytes, may exert an active role in the pathogenesis of TBI. Astrocytes, the predominant cell type in the human brain, are traditionally associated with providing only structural support within the CNS. However, recent work suggests astrocytes may regulate brain homeostasis and limit brain injury. In contrast, reactive astrocytes may also contribute to increased neuroinflammation, the development of cerebral edema, and elevated intracranial pressure, suggesting possible roles in exacerbating secondary brain injury following neurotrauma. The multiple, opposing roles for astrocytes following neurotrauma may have important implications for the design of directed therapeutics to limit neurological injury. As such, a primary focus of this review is to summarize the emerging evidence suggesting reactive astrocytes influence the response of the brain to TBI.


Subject(s)
Astrocytes/metabolism , Astrocytes/pathology , Brain Injuries/metabolism , Brain Injuries/pathology , Animals , Brain/metabolism , Brain/pathology , Brain Edema/metabolism , Brain Edema/pathology , Brain Edema/therapy , Brain Injuries/therapy , Humans , Neurons/metabolism , Neurons/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...