Network


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

Hotspot


Dive into the research topics where Susan C. Buckingham is active.

Publication


Featured researches published by Susan C. Buckingham.


Nature Medicine | 2011

Glutamate release by primary brain tumors induces epileptic activity

Susan C. Buckingham; Susan L. Campbell; Brian R. Haas; Vedrana Montana; Stefanie Robel; Toyin Ogunrinu; Harald Sontheimer

Epileptic seizures are a common and poorly understood comorbidity for individuals with primary brain tumors. To investigate peritumoral seizure etiology, we implanted human-derived glioma cells into severe combined immunodeficient mice. Within 14–18 d, glioma-bearing mice developed spontaneous and recurring abnormal electroencephalogram events consistent with progressive epileptic activity. Acute brain slices from these mice showed marked glutamate release from the tumor mediated by the system xc− cystine-glutamate transporter (encoded by Slc7a11). Biophysical and optical recordings showed glutamatergic epileptiform hyperexcitability that spread into adjacent brain tissue. We inhibited glutamate release from the tumor and the ensuing hyperexcitability by sulfasalazine (SAS), a US Food and Drug Administration–approved drug that blocks system xc−. We found that acute administration of SAS at concentrations equivalent to those used to treat Crohns disease in humans reduced epileptic event frequency in tumor-bearing mice compared with untreated controls. SAS should be considered as an adjuvant treatment to ameliorate peritumoral seizures associated with glioma in humans.


The Journal of Neuroscience | 2015

Reactive Astrogliosis Causes the Development of Spontaneous Seizures

Stefanie Robel; Susan C. Buckingham; Jessica L. Boni; Susan L. Campbell; Niels C. Danbolt; Therese Riedemann; Bernd Sutor; Harald Sontheimer

Epilepsy is one of the most common chronic neurologic diseases, yet approximately one-third of affected patients do not respond to anticonvulsive drugs that target neurons or neuronal circuits. Reactive astrocytes are commonly found in putative epileptic foci and have been hypothesized to be disease contributors because they lose essential homeostatic capabilities. However, since brain pathology induces astrocytes to become reactive, it is difficult to distinguish whether astrogliosis is a cause or a consequence of epileptogenesis. We now present a mouse model of genetically induced, widespread chronic astrogliosis after conditional deletion of β1-integrin (Itgβ1). In these mice, astrogliosis occurs in the absence of other pathologies and without BBB breach or significant inflammation. Electroencephalography with simultaneous video recording revealed that these mice develop spontaneous seizures during the first six postnatal weeks of life and brain slices show neuronal hyperexcitability. This was not observed in mice with neuronal-targeted β1-integrin deletion, supporting the hypothesis that astrogliosis is sufficient to induce epileptic seizures. Whole-cell patch-clamp recordings from astrocytes further suggest that the heightened excitability was associated with impaired astrocytic glutamate uptake. Moreover, the relative expression of the cation-chloride cotransporters (CCC) NKCC1 (Slc12a2) and KCC2 (Slc12a5), which are responsible for establishing the neuronal Cl− gradient that governs GABAergic inhibition were altered and the NKCC1 inhibitor bumetanide eliminated seizures in a subgroup of mice. These data suggest that a shift in the relative expression of neuronal NKCC1 and KCC2, similar to that observed in immature neurons during development, may contribute to astrogliosis-associated seizures.


Science Translational Medicine | 2015

SLC7A11 expression is associated with seizures and predicts poor survival in patients with malignant glioma

Stephanie M. Robert; Susan C. Buckingham; Susan L. Campbell; Stefanie Robel; Kenneth T. Holt; Toyin Ogunrinu-Babarinde; Paula Warren; David M. White; Meredith A. Reid; Jenny M. Eschbacher; Michael E. Berens; Adrienne C. Lahti; Louis B. Nabors; Harald Sontheimer

SLC7A11, the catalytic subunit of the cystine/glutamate antiporter, System xc− (SXC), is up-regulated in a subpopulation of patient gliomas, where it is responsible for excitotoxic glutamate release, accelerated tumor growth, and tumor-associated seizures. Seizing an opportunity to study glioma Gliomas are the most common type of malignant brain tumors, and they frequently cause seizures. A new study by Robert et al. uncovers some of the mechanisms involved in this process, showing how a specific cystine/glutamate transporter contributes to excitotoxic glutamate release, causing the death of surrounding cells and inducing seizures. The authors also showed that tumors expressing this transporter were more aggressive and grew more quickly, possibly because the destruction of surrounding normal cells allowed the tumors to expand more rapidly. These findings suggest that the expression of this cystine/glutamate transporter may be useful as a predictor of outcome and a potential therapeutic target in glioma. Glioma is the most common malignant primary brain tumor. Its rapid growth is aided by tumor-mediated glutamate release, creating peritumoral excitotoxic cell death and vacating space for tumor expansion. Glioma glutamate release may also be responsible for seizures, which complicate the clinical course for many patients and are often the presenting symptom. A hypothesized glutamate release pathway is the cystine/glutamate transporter System xc− (SXC), responsible for the cellular synthesis of glutathione (GSH). However, the relationship of SXC-mediated glutamate release, seizures, and tumor growth remains unclear. Probing expression of SLC7A11/xCT, the catalytic subunit of SXC, in patient and mouse-propagated tissues, we found that ~50% of patient tumors have elevated SLC7A11 expression. Compared with tumors lacking this transporter, in vivo propagated and intracranially implanted SLC7A11-expressing tumors grew faster, produced pronounced peritumoral glutamate excitotoxicity, induced seizures, and shortened overall survival. In agreement with animal data, increased SLC7A11 expression predicted shorter patient survival according to genomic data in the REMBRANDT (National Institutes of Health Repository for Molecular Brain Neoplasia Data) database. In a clinical pilot study, we used magnetic resonance spectroscopy to determine SXC-mediated glutamate release by measuring acute changes in glutamate after administration of the U.S. Food and Drug Administration–approved SXC inhibitor, sulfasalazine (SAS). In nine glioma patients with biopsy-confirmed SXC expression, we found that expression positively correlates with glutamate release, which is acutely inhibited with oral SAS. These data suggest that SXC is the major pathway for glutamate release from gliomas and that SLC7A11 expression predicts accelerated growth and tumor-associated seizures.


The Journal of Neuroscience | 2015

Tau-dependent Kv4.2 depletion and dendritic hyperexcitability in a mouse model of Alzheimer's disease.

Alicia M. Hall; Benjamin T. Throesch; Susan C. Buckingham; Sean J. Markwardt; Yin Peng; Qin Wang; Dax A. Hoffman; Erik D. Roberson

Neuronal hyperexcitability occurs early in the pathogenesis of Alzheimers disease (AD) and contributes to network dysfunction in AD patients. In other disorders with neuronal hyperexcitability, dysfunction in the dendrites often contributes, but dendritic excitability has not been directly examined in AD models. We used dendritic patch-clamp recordings to measure dendritic excitability in the CA1 region of the hippocampus. We found that dendrites, more so than somata, of hippocampal neurons were hyperexcitable in mice overexpressing Aβ. This dendritic hyperexcitability was associated with depletion of Kv4.2, a dendritic potassium channel important for regulating dendritic excitability and synaptic plasticity. The antiepileptic drug, levetiracetam, blocked Kv4.2 depletion. Tau was required, as crossing with tau knock-out mice also prevented both Kv4.2 depletion and dendritic hyperexcitability. Dendritic hyperexcitability induced by Kv4.2 deficiency exacerbated behavioral deficits and increased epileptiform activity in hAPP mice. We conclude that increased dendritic excitability, associated with changes in dendritic ion channels including Kv4.2, may contribute to neuronal dysfunction in early stages AD.


Glia | 2015

GABAergic disinhibition and impaired KCC2 cotransporter activity underlie tumor-associated epilepsy

Susan L. Campbell; Stefanie Robel; Vishnu Anand Cuddapah; Stephanie M. Robert; Susan C. Buckingham; Kristopher T. Kahle; Harald Sontheimer

Seizures frequently accompany gliomas and often escalate to peritumoral epilepsy. Previous work revealed the importance of tumor‐derived excitatory glutamate (Glu) release mediated by the cystine‐glutamate transporter (SXC) in epileptogenesis. We now show a novel contribution of GABAergic disinhibition to disease pathophysiology. In a validated mouse glioma model, we found that peritumoral parvalbumin‐positive GABAergic inhibitory interneurons are significantly reduced, corresponding with deficits in spontaneous and evoked inhibitory neurotransmission. Most remaining peritumoral neurons exhibit elevated intracellular Cl− concentration ([Cl−]i) and consequently depolarizing, excitatory gamma‐aminobutyric acid (GABA) responses. In these neurons, the plasmalemmal expression of KCC2, which establishes the low [Cl−]i required for GABAAR‐mediated inhibition, is significantly decreased. Interestingly, reductions in inhibition are independent of Glu release, but the presence of both decreased inhibition and decreased SXC expression is required for epileptogenesis. We suggest GABAergic disinhibition renders peritumoral neuronal networks hyper‐excitable and susceptible to seizures triggered by excitatory stimuli, and propose KCC2 as a therapeutic target. GLIA 2015;63:23–36


Neuroscience | 2013

STATUS EPILEPTICUS TRIGGERS EARLY AND LATE ALTERATIONS IN BRAIN-DERIVED NEUROTROPHIC FACTOR AND NMDA GLUTAMATE RECEPTOR GRIN2B DNA METHYLATION LEVELS IN THE HIPPOCAMPUS

R. Ryley Parrish; Asher J. Albertson; Susan C. Buckingham; John J. Hablitz; Katherine L. Mascia; W. Davis Haselden; Farah D. Lubin

Status epilepticus (SE) triggers abnormal expression of genes in the hippocampus, such as glutamate receptor subunit epsilon-2 (Grin2b/Nr2b) and brain-derived neurotrophic factor (Bdnf), that is thought to occur in temporal lobe epilepsy (TLE). We examined the underlying DNA methylation mechanisms and investigated whether these mechanisms contribute to the expression of these gene targets in the epileptic hippocampus. Experimental TLE was provoked by kainic acid-induced SE. Bisulfite sequencing analysis revealed increased Grin2b/Nr2b and decreased Bdnf DNA methylation levels that corresponded to decreased Grin2b/Nr2b and increased Bdnf mRNA and protein expression in the epileptic hippocampus. Blockade of DNA methyltransferase (DNMT) activity with zebularine decreased global DNA methylation levels and reduced Grin2b/Nr2b, but not Bdnf, DNA methylation levels. Interestingly, we found that DNMT blockade further decreased Grin2b/Nr2b mRNA expression whereas GRIN2B protein expression increased in the epileptic hippocampus, suggesting that a posttranscriptional mechanism may be involved. Using chromatin immunoprecipitation analysis we found that DNMT inhibition restored the decreases in AP2alpha transcription factor levels at the Grin2b/Nr2b promoter in the epileptic hippocampus. DNMT inhibition increased field excitatory postsynaptic potential in hippocampal slices isolated from epileptic rats. Electroencephalography (EEG) monitoring confirmed that DNMT inhibition did not significantly alter the disease course, but promoted the latency to seizure onset or SE. Thus, DNA methylation may be an early event triggered by SE that persists late into the epileptic hippocampus to contribute to gene expression changes in TLE.


Epilepsia | 2012

Human glioma cells induce hyperexcitability in cortical networks

Susan L. Campbell; Susan C. Buckingham; Harald Sontheimer

Purpose:  Patients with gliomas frequently present with seizures, but the factors associated with seizure development are still poorly understood. In this study, we assessed peritumoral synaptic network activity in a glioma animal model and tested the contribution of aberrant glutamate release from gliomas on glioma‐associated epileptic network activity.


Neurochemistry International | 2013

Glutamate and tumor-associated epilepsy: glial cell dysfunction in the peritumoral environment

Susan C. Buckingham; Stefanie Robel

Seizures are a serious and debilitating co-morbidity of primary brain tumors that affect most patients, yet their etiology is poorly understood. In many CNS pathologies, including epilepsy and brain injury, high levels of extracellular glutamate have been implicated in seizure generation. It has been shown that gliomas release neurotoxic levels of glutamate through their high expression of system xc-. More recently it was shown that the surrounding peritumoral cortex is spontaneously hyperexcitable. In this review, we discuss how gliomas induce changes in the surrounding environment that may further contribute to elevated extracellular glutamate and tumor-associated seizures. Peritumoral astrocytes become reactive and lose their ability to remove glutamate, while microglia, in response to signals from glioma cells, may release glutamate. In addition, gliomas increase blood brain barrier permeability, allowing seizure-inducing serum components, including glutamate, into the peritumoral region. These factors, working together or alone, may influence the frequency and severity of tumor-associated epilepsy.


Annals of clinical and translational neurology | 2015

Methionine increases BDNF DNA methylation and improves memory in epilepsy

R. Ryley Parrish; Susan C. Buckingham; Katherine L. Mascia; Jarvis J. Johnson; Michal M. Matyjasik; Roxanne M. Lockhart; Farah D. Lubin

Temporal lobe epilepsy (TLE) patients exhibit signs of memory impairments even when seizures are pharmacologically controlled. Surprisingly, the underlying molecular mechanisms involved in TLE‐associated memory impairments remain elusive. Memory consolidation requires epigenetic transcriptional regulation of genes in the hippocampus; therefore, we aimed to determine how epigenetic DNA methylation mechanisms affect learning‐induced transcription of memory‐permissive genes in the epileptic hippocampus.


Epilepsia | 2014

Complement C5‐deficient mice are protected from seizures in experimental cerebral malaria

Susan C. Buckingham; Theresa N. Ramos; Scott R. Barnum

Studies have demonstrated that the membrane attack complex (MAC) of complement can evoke seizures when injected directly into rodent brain. In the course of studies that examine the role of complement in the development of experimental cerebral malaria (ECM), we observed fewer seizures in mice deficient in C5, a component required for MAC formation. To determine if the MAC contributed to the tonic–clonic seizures characteristic of ECM, we performed long‐term video–electroencephalography (EEG) on C5−/− mice with Plasmodium berghei ANKA‐induced cerebral malaria and observed significantly reduced spike and seizure frequency compared to wild‐type mice. Our data suggest a role for the MAC in malaria‐induced seizures and that inhibition of the terminal complement pathway may reduce seizures and seizure‐related neurocognitive deficits.

Collaboration


Dive into the Susan C. Buckingham's collaboration.

Top Co-Authors

Avatar

Harald Sontheimer

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Susan L. Campbell

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Stefanie Robel

Ludwig Maximilian University of Munich

View shared research outputs
Top Co-Authors

Avatar

Erik D. Roberson

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Farah D. Lubin

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Alicia M. Hall

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Benjamin T. Throesch

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Dax A. Hoffman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jessica L. Boni

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

John J. Hablitz

Baylor College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge