Network


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

Hotspot


Dive into the research topics where Bruce Mickey is active.

Publication


Featured researches published by Bruce Mickey.


Nature Medicine | 2012

2-hydroxyglutarate detection by magnetic resonance spectroscopy in IDH -mutated patients with gliomas

Changho Choi; Sandeep K. Ganji; Ralph J. DeBerardinis; Kimmo J. Hatanpaa; Dinesh Rakheja; Zoltan Kovacs; Xiao Li Yang; Tomoyuki Mashimo; Jack Raisanen; Isaac Marin-Valencia; Juan M. Pascual; Christopher Madden; Bruce Mickey; Craig R. Malloy; Robert M. Bachoo; Elizabeth A. Maher

Mutations in isocitrate dehydrogenases 1 and 2 (IDH1 and IDH2) have been shown to be present in most World Health Organization grade 2 and grade 3 gliomas in adults. These mutations are associated with the accumulation of 2-hydroxyglutarate (2HG) in the tumor. Here we report the noninvasive detection of 2HG by proton magnetic resonance spectroscopy (MRS). We developed and optimized the pulse sequence with numerical and phantom analyses for 2HG detection, and we estimated the concentrations of 2HG using spectral fitting in the tumors of 30 subjects. Detection of 2HG correlated with mutations in IDH1 or IDH2 and with increased levels of D-2HG by mass spectrometry of the resected tumors. Noninvasive detection of 2HG may prove to be a valuable diagnostic and prognostic biomarker.


Cell Metabolism | 2012

Analysis of tumor metabolism reveals mitochondrial glucose oxidation in genetically diverse, human glioblastomas in the mouse brain in vivo

Isaac Marin-Valencia; Chendong Yang; Tomoyuki Mashimo; Steve K. Cho; Hyeonman Baek; Xiao Li Yang; Kartik N. Rajagopalan; Melissa Maddie; Vamsidhara Vemireddy; Zhenze Zhao; Ling Cai; Levi B. Good; Benjamin P. Tu; Kimmo J. Hatanpaa; Bruce Mickey; José M. Matés; Juan M. Pascual; Elizabeth A. Maher; Craig R. Malloy; Ralph J. DeBerardinis; Robert M. Bachoo

Dysregulated metabolism is a hallmark of cancer cell lines, but little is known about the fate of glucose and other nutrients in tumors growing in their native microenvironment. To study tumor metabolism in vivo, we used an orthotopic mouse model of primary human glioblastoma (GBM). We infused (13)C-labeled nutrients into mice bearing three independent GBM lines, each with a distinct set of mutations. All three lines displayed glycolysis, as expected for aggressive tumors. They also displayed unexpected metabolic complexity, oxidizing glucose via pyruvate dehydrogenase and the citric acid cycle, and using glucose to supply anaplerosis and other biosynthetic activities. Comparing the tumors to surrounding brain revealed obvious metabolic differences, notably the accumulation of a large glutamine pool within the tumors. Many of these same activities were conserved in cells cultured ex vivo from the tumors. Thus GBM cells utilize mitochondrial glucose oxidation during aggressive tumor growth in vivo.


Cancer Research | 2009

EGFRvIII and DNA Double-Strand Break Repair: A Molecular Mechanism for Radioresistance in Glioblastoma

Bipasha Mukherjee; Brian McEllin; Cristel V. Camacho; Nozomi Tomimatsu; Shyam Sirasanagandala; Suraj Nannepaga; Kimmo J. Hatanpaa; Bruce Mickey; Christopher Madden; Elizabeth A. Maher; David A. Boothman; Frank B. Furnari; Webster K. Cavenee; Robert M. Bachoo; Sandeep Burma

Glioblastoma multiforme (GBM) is the most lethal of brain tumors and is highly resistant to ionizing radiation (IR) and chemotherapy. Here, we report on a molecular mechanism by which a key glioma-specific mutation, epidermal growth factor receptor variant III (EGFRvIII), confers radiation resistance. Using Ink4a/Arf-deficient primary mouse astrocytes, primary astrocytes immortalized by p53/Rb suppression, as well as human U87 glioma cells, we show that EGFRvIII expression enhances clonogenic survival following IR. This enhanced radioresistance is due to accelerated repair of DNA double-strand breaks (DSB), the most lethal lesion inflicted by IR. The EGFR inhibitor gefitinib (Iressa) and the phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 attenuate the rate of DSB repair. Importantly, expression of constitutively active, myristylated Akt-1 accelerates repair, implicating the PI3K/Akt-1 pathway in radioresistance. Most notably, EGFRvIII-expressing U87 glioma cells show elevated activation of a key DSB repair enzyme, DNA-dependent protein kinase catalytic subunit (DNA-PKcs). Enhanced radioresistance is abrogated by the DNA-PKcs-specific inhibitor NU7026, and EGFRvIII fails to confer radioresistance in DNA-PKcs-deficient cells. In vivo, orthotopic U87-EGFRvIII-derived tumors display faster rates of DSB repair following whole-brain radiotherapy compared with U87-derived tumors. Consequently, EGFRvIII-expressing tumors are radioresistant and continue to grow following whole-brain radiotherapy with little effect on overall survival. These in vitro and in vivo data support our hypothesis that EGFRvIII expression promotes DNA-PKcs activation and DSB repair, perhaps as a consequence of hyperactivated PI3K/Akt-1 signaling. Taken together, our results raise the possibility that EGFR and/or DNA-PKcs inhibition concurrent with radiation may be an effective therapeutic strategy for radiosensitizing high-grade gliomas.


Clinical Cancer Research | 2010

The Telomerase Antagonist, Imetelstat, Efficiently Targets Glioblastoma Tumor-Initiating Cells Leading to Decreased Proliferation and Tumor Growth

Calin O. Marian; Steve K. Cho; Brian McEllin; Elizabeth A. Maher; Kimmo J. Hatanpaa; Christopher Madden; Bruce Mickey; Woodring E. Wright; Jerry W. Shay; Robert M. Bachoo

Purpose: Telomerase activity is one of the hallmarks of cancer and is a highly relevant therapeutic target. The effects of a novel human telomerase antagonist, imetelstat, on primary human glioblastoma (GBM) tumor-initiating cells were investigated in vitro and in vivo. Experimental Design: Tumor-initiating cells were isolated from primary GBM tumors and expanded as neurospheres in vitro. The GBM tumor-initiating cells were treated with imetelstat and examined for the effects on telomerase activity levels, telomere length, proliferation, clonogenicity, and differentiation. Subsequently, mouse orthotopic and subcutaneous xenografts were used to assess the in vivo efficacy of imetelstat. Results: Imetelstat treatment produced a dose-dependent inhibition of telomerase (IC50 0.45 μmol/L). Long-term imetelstat treatment led to progressive telomere shortening, reduced rates of proliferation, and eventually cell death in GBM tumor-initiating cells. Imetelstat in combination with radiation and temozolomide had a dramatic effect on cell survival and activated the DNA damage response pathway. Imetelstat is able to cross the blood-brain barrier in orthotopic GBM xenograft tumors. Fluorescently labeled GBM tumor cells isolated from orthotopic tumors, following systemic administration of imetelstat (30 mg/kg every day for three days), showed ∼70% inhibition of telomerase activity. Chronic systemic treatment produced a marked decrease in the rate of xenograft subcutaneous tumor growth. Conclusion: This preclinical study supports the feasibility of testing imetelstat in the treatment of GBM patients, alone or in combination with standard therapies. Clin Cancer Res; 16(1); 154–63


NMR in Biomedicine | 2012

Metabolism of [U-13 C]glucose in human brain tumors in vivo.

Elizabeth A. Maher; Isaac Marin-Valencia; Robert M. Bachoo; Tomoyuki Mashimo; Jack Raisanen; Kimmo J. Hatanpaa; Ashish Jindal; F. Mark Jeffrey; Changho Choi; Christopher Madden; Dana Mathews; Juan M. Pascual; Bruce Mickey; Craig R. Malloy; Ralph J. DeBerardinis

Glioblastomas and brain metastases demonstrate avid uptake of 2‐[18F]fluoro‐2‐deoxyglucose by positron emission tomography and display perturbations of intracellular metabolite pools by 1H MRS. These observations suggest that metabolic reprogramming contributes to brain tumor growth in vivo. The Warburg effect, excess metabolism of glucose to lactate in the presence of oxygen, is a hallmark of cancer cells in culture. 2‐[18F]Fluoro‐2‐deoxyglucose‐positive tumors are assumed to metabolize glucose in a similar manner, with high rates of lactate formation relative to mitochondrial glucose oxidation, but few studies have specifically examined the metabolic fates of glucose in vivo. In particular, the capacity of human brain cancers to oxidize glucose in the tricarboxylic acid cycle is unknown. Here, we studied the metabolism of human brain tumors in situ. [U‐13 C]Glucose (uniformly labeled glucose, i.e. d‐glucose labeled with 13 C in all six carbons) was infused during surgical resection, and tumor samples were subsequently subjected to 13C NMR spectroscopy. The analysis of tumor metabolites revealed lactate production, as expected. We also determined that pyruvate dehydrogenase, turnover of the tricarboxylic acid cycle, anaplerosis and de novo glutamine and glycine synthesis contributed significantly to the ultimate disposition of glucose carbon. Surprisingly, less than 50% of the acetyl‐coenzyme A pool was derived from blood‐borne glucose, suggesting that additional substrates contribute to tumor bioenergetics. This study illustrates a convenient approach that capitalizes on the high information content of 13C NMR spectroscopy and enables the analysis of intermediary metabolism in diverse cancers growing in their native microenvironment. Copyright


Proceedings of the National Academy of Sciences of the United States of America | 2014

In vivo chemical exchange saturation transfer imaging allows early detection of a therapeutic response in glioblastoma

Koji Sagiyama; Tomoyuki Mashimo; Osamu Togao; Vamsidhara Vemireddy; Kimmo J. Hatanpaa; Elizabeth A. Maher; Bruce Mickey; Edward Pan; A. Dean Sherry; Robert M. Bachoo; Masaya Takahashi

Significance The prognosis and management of patients with glioma is vastly different depending on whether one detects tumor progression or treatment effects. Although the gold standard in the evaluation of treatment efficacy involves MRI, the currently available imaging methods often do not suffice to make the final decision. Our study demonstrated that amide proton transfer (APT) imaging, one subset of chemical exchange saturation transfer imaging, can detect molecular signals in glioma induced by short-term chemotherapy with temozolomide. These molecular events precede morphologic changes. The APT signal did not decrease in tumors resistant to chemotherapy. APT imaging may provide a useful prognostic biomarker of treatment response or tumor progression in glioma. Glioblastoma multiforme (GBM), which account for more than 50% of all gliomas, is among the deadliest of all human cancers. Given the dismal prognosis of GBM, it would be advantageous to identify early biomarkers of a response to therapy to avoid continuing ineffective treatments and to initiate other therapeutic strategies. The present in vivo longitudinal study in an orthotopic mouse model demonstrates quantitative assessment of early treatment response during short-term chemotherapy with temozolomide (TMZ) by amide proton transfer (APT) imaging. In a GBM line, only one course of TMZ (3 d exposure and 4 d rest) at a dose of 80 mg/kg resulted in substantial reduction in APT signal compared with untreated control animals, in which the APT signal continued to increase. Although there were no detectable differences in tumor volume, cell density, or apoptosis rate between groups, levels of Ki67 (index of cell proliferation) were substantially reduced in treated tumors. In another TMZ-resistant GBM line, the APT signal and levels of Ki67 increased despite the same course of TMZ treatment. As metabolite changes are known to occur early in the time course of chemotherapy and precede morphologic changes, these results suggest that the APT signal in glioma may be a useful functional biomarker of treatment response or degree of tumor progression. Thus, APT imaging may serve as a sensitive biomarker of early treatment response and could potentially replace invasive biopsies to provide a definitive diagnosis. This would have a major impact on the clinical management of patients with glioma.


Otolaryngology-Head and Neck Surgery | 1992

Complications of lumbar spinal fluid drainage.

Peter S. Roland; Brad Marple; William L. Meyerhoff; Bruce Mickey

Cerebrospinal fluid fistula is an unfortunate, yet well-recognized, complication of basilar skull fracture, skull base surgery, and neurotologic procedures. Treatment commonly involves the use of continuous lumbar drainage. A retrospective chart review of 32 consecutive patients who required placement of lumbar drain by the otorhinolaryngology and neurosurgical services from March 1988 through July 1991 was undertaken to assess possible complications. The complications found were readily separated into minor and major categories on the basis of the possibility of permanent morbidity or mortality. Minor complications, including subjective complaints of headache, nausea, vomiting, etc., were noted in 59% of patients. Major complications were observed in four of 32 patients (12.5%), including unilateral occlusion of the posterior cerebral artery and unilateral true vocal cord paralysis. Alleviation of all complications was achieved by cessation of lumbar drainage. These cases are presented with discussion of pathogenesis. These findings demonstrate the possibility of potentially serious complications that mandate close monitoring of patients who require continuous lumbar drainage.


Journal of Clinical Anesthesia | 2000

Use of the laryngeal mask airway during awake craniotomy for tumor resection

W. Kendall Tongier; Girish P. Joshi; Dennis F. Landers; Bruce Mickey

There is an increasing trend toward performing craniotomy for primary brain tumor excision with local anesthesia. We report the use of the laryngeal mask airway as a part of an anesthetic technique designed for patients requiring awake cortical mapping during brain tumor excision.


Neurosurgery | 1998

Surgical Treatment of Cerebrospinal Fluid Fistulae Involving Lateral Extension of the Sphenoid Sinus

Fraser Landreneau; Bruce Mickey; Caetano Coimbra

OBJECTIVE AND IMPORTANCE Four cases of spontaneous cerebrospinal fluid rhinorrhea caused by communication between the subarachnoid space of the middle cranial fossa and a lateral extension of the sphenoid sinus are presented. The cause and management of this unique type of cranial base defect are discussed. CLINICAL PRESENTATION During the past 10 years, four patients referred to our institution with atraumatic cerebrospinal fluid fistulae were observed to have temporal encephaloceles (encephalomeningoceles) traversing the floor of the middle cranial fossa. Three of the patients had previously undergone unsuccessful transnasal attempts to repair their fistulae by obliteration of the sphenoid sinus. The fourth patient presented before undergoing any treatment. No patient had associated hydrocephalus or tumor. Preoperative computed tomographic cisternograms revealed that all fistulae involved a lateral extension of the sphenoid sinus into the floor of the middle cranial fossa. INTERVENTION After definitive localization, each patient was operated on transcranially through an anterior middle cranial fossa approach with extradural and/or intradural exploration. The associated temporal encephalocele was amputated or disconnected, and the dehiscent dura and middle cranial fossa floor defect were oversewn and packed with autogenous tissue, respectively. CONCLUSION The surgical treatment of cerebrospinal fluid rhinorrhea secondary to middle fossa encephalocele associated with lateral extension of the sphenoidal sinus differs from the surgical strategy for more medial sphenoidal fistulae. Fistulae involving a lateral extension of the sphenoid sinus require a transcranial approach for direct visualization and obliteration of the defect, whereas fistulae involving the central portion of the sinus may be successfully obliterated transsphenoidally.


Cancer Research | 2009

The Receptor Interacting Protein 1 Inhibits p53 Induction through NF-κB Activation and Confers a Worse Prognosis in Glioblastoma

Seongmi Park; Kimmo J. Hatanpaa; Yang Xie; Bruce Mickey; Christopher Madden; Jack Raisanen; Deepti B. Ramnarain; Guanghua Xiao; Debabrata Saha; David A. Boothman; Dawen Zhao; Robert M. Bachoo; Russell O. Pieper; Amyn A. Habib

Nuclear factor-kappaB (NF-kappaB) activation may play an important role in the pathogenesis of cancer and also in resistance to treatment. Inactivation of the p53 tumor suppressor is a key component of the multistep evolution of most cancers. Links between the NF-kappaB and p53 pathways are under intense investigation. In this study, we show that the receptor interacting protein 1 (RIP1), a central component of the NF-kappaB signaling network, negatively regulates p53 tumor suppressor signaling. Loss of RIP1 from cells results in augmented induction of p53 in response to DNA damage, whereas increased RIP1 level leads to a complete shutdown of DNA damage-induced p53 induction by enhancing levels of cellular mdm2. The key signal generated by RIP1 to up-regulate mdm2 and inhibit p53 is activation of NF-kappaB. The clinical implication of this finding is shown in glioblastoma, the most common primary malignant brain tumor in adults. We show that RIP1 is commonly overexpressed in glioblastoma, but not in grades II and III glioma, and increased expression of RIP1 confers a worse prognosis in glioblastoma. Importantly, RIP1 levels correlate strongly with mdm2 levels in glioblastoma. Our results show a key interaction between the NF-kappaB and p53 pathways that may have implications for the targeted treatment of glioblastoma.

Collaboration


Dive into the Bruce Mickey's collaboration.

Top Co-Authors

Avatar

Kimmo J. Hatanpaa

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Elizabeth A. Maher

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Robert M. Bachoo

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Christopher Madden

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Samuel L. Barnett

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Tomoyuki Mashimo

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Changho Choi

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Craig R. Malloy

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jack Raisanen

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ralph J. DeBerardinis

University of Texas Southwestern Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge